EP4183802A1 - Anti-ang-2-antikörper und verwendung davon - Google Patents

Anti-ang-2-antikörper und verwendung davon Download PDF

Info

Publication number
EP4183802A1
EP4183802A1 EP21827966.9A EP21827966A EP4183802A1 EP 4183802 A1 EP4183802 A1 EP 4183802A1 EP 21827966 A EP21827966 A EP 21827966A EP 4183802 A1 EP4183802 A1 EP 4183802A1
Authority
EP
European Patent Office
Prior art keywords
antibody
seq
amino acid
acid sequence
sequence shown
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21827966.9A
Other languages
English (en)
French (fr)
Inventor
Yiming Li
Li Li
Fenggen FU
Hua JING
Junjian Liu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Innovent Biologics Suzhou Co Ltd
Original Assignee
Innovent Biologics Suzhou Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Innovent Biologics Suzhou Co Ltd filed Critical Innovent Biologics Suzhou Co Ltd
Publication of EP4183802A1 publication Critical patent/EP4183802A1/de
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to novel antibodies and antibody fragments thereof, which specifically bind to ANG-2, and to compositions comprising the antibodies or antibody fragments thereof.
  • the present invention relates to nucleic acids encoding the antibodies or antibody fragments thereof and host cells containing the nucleic acids, and their uses.
  • the present invention relates to therapeutic and diagnostic uses of the antibodies and antibody fragments.
  • Angiopoietin-2 also known as ANGPT2 or Ang2 is a member of the angiopoietin family. This family consists of four members, including Ang1, Ang2, and Ang3/4. Angiopoietins Ang1 and Ang2, as the ligands of Tie2 receptor (tyrosine kinase with Ig and EGF homology domains, also known as TEK), competitively bind to Tie2 expressed on the surface of vascular endothelial cells and are involved in a variety of blood vessel-related physiological and pathological processes.
  • Tie2 receptor tyrosine kinase with Ig and EGF homology domains, also known as TEK
  • Ang1 is expressed by vascular pericytes and its binding to Tie2 causes Tie2 phosphorylation and signaling pathway activation. Thus, it plays a role in maintaining the structural and functional integrity of mature blood vessels.
  • Ang-2 a natural antagonist of Ang-1, is believed to play a role in destabilizing blood vessels by antagonizing the ANG-1/Tie2 signaling pathway.
  • Peters S. et al. showed that, the permeability of endothelial cells in vitro was increased when ang-2 or both ang-2 and VEGF were added to the culture medium containing endothelial cells ( Peters S.
  • vascular permeability and angiogenesis Several diseases have been known to be associated with vascular permeability and angiogenesis, including inflammation, pneumonia, mycoplasma pneumonia infection, sepsis, solid tumors, and ocular fundus diseases.
  • Preclinical studies using an acute ocular inflammation model and a choroidal neovascularization model (CNV model) have shown that ang-2 and VEGF are synergistically involved in the pathological neovascularization and vascular permeability. Therefore, in addition to anti-VEGF therapy, the Tie2/angiopoietin signaling pathway has also been proposed as a therapeutic target for vascular-related retinopathies such as neovascular age-related macular degeneration (nAMD) and diabetic macular edema (DME).
  • nAMD neovascular age-related macular degeneration
  • DME diabetic macular edema
  • Nesvacumab (Regeneron, Tarrytown, NY, USA), an IgG1-type monoclonal antibody that selectively binds to ANG2 with high affinity and specifically blocks ANG2 binding to Tie2 receptor, was proposed for the treatment of nAMD and DME in combination with aflibercept (anti-VEGF-A).
  • aflibercept anti-VEGF-A
  • Faricimab a bispecific antibody that inhibits both VEGF-A and ANG2 (developed by Roche, Basel, Switzerland and Genentech, South San Francisco, CA, USA), was proposed for the treatment of nAMD and DME.
  • the currently available therapies still have limitations in terms of efficacy and treatment burden. See, Rehan M.
  • Pulmonary inflammatory conditions including acute bacterial or viral pneumonia and acute respiratory distress syndrome (ARDS) and acute lung injury, are characterized by leukocyte accumulation and increased microvascular permeability.
  • ARDS has attracted much attention due to its association with severe lung injury and high mortality.
  • ARDS manifests severe hypoxemia, decreased lung compliance, diffuse alveolar damage, and bilateral lung infiltration.
  • ARDS can be caused by direct or indirect lung injury, in which the inflammation in the lung induces the damage to alveolar-capillary barrier, which is one of the significant and direct factors that contributes to ARDS.
  • the current main treatment modality for ARDS patients is mechanical ventilation.
  • non-mechanical ventilation treatment modalities such as lung water removal and fluid management, surfactant replenishment, beta-receptor agonists, statins, and glucocorticoids, but for most of them, reliable efficacy has not been demonstrated.
  • the efficacy of the hormone drug dexamethasone in the treatment of inflammation-mediated lung injury has been clinically recognized (DOI: 10.1056/NEJMoa2021436)
  • the drug has enormous side effects, and the profound impacts of the drug on the subsequent life of patients have been increasingly reported.
  • ANG-2 in vascular permeability and angiogenesis, there is a need in the field for continuous development of new drug molecules that are capable of highly specifically and selectively targeting and regulating the biological activity of ANG-2, such as anti-ANG-2 antibodies and combination therapies thereof, for the treatment of vascular-related diseases.
  • the present invention provides anti-ANG-2 antibodies, their encoding genes, and uses thereof. Based on hybridoma screening, chimeric antibody construction, humanization and affinity maturation, the inventors have developed anti-human ANG-2 antibodies of the invention with high affinity and specificity.
  • the antibodies of the invention can effectively block the ANG-2/receptor TIE2 signaling pathway and the ANG-2/integrin signaling pathway, and work synergistically with anti-VEGF agents to reduce vascular leakage, inhibite neovascularization and improve vascular integrity in an animal model of choroidal neovascularization.
  • the antibodies of the invention exhibit low non-specific binding to a variety of retinal-derived cells, thereby facilitating its intravitreous administration at high concentrations for the treatment of ocular fundus diseases.
  • the anti-ANG2 antibodies of the invention can improve vascular permeability, vascular leakage and vascular inflammatory conditions, and ameliorate various parameters in pulmonary inflammatory conditions, especially ARDS, such as pulmonary infiltration of inflammatory cells, blood oxygen saturation, pulmonary edema degree, lung weight index, and animal survival rate.
  • ARDS pulmonary infiltration of inflammatory cells
  • the treatment methods with the anti-ANG2 antibdoies of the invention are hormone-independent and can achieve an efficacy that is not inferior to, and in some parameters even superior to, that of the hormone dexamethasone.
  • the present invention provides novel anti-Ang-2 antibodies, and antigen-binding fragments thereof.
  • the anti-ANG-2 antibodies of the invention exhibite one or more of the following properties:
  • the present invention provides an antibody or antigen-binding fragment thereof that binds to ANG-2, comprising the HCDR1, HCDR2 and HCDR3 sequences of one of the heavy chain variable regions shown in SEQ ID NOs: 19, 21, 23, 25, 27, and 29, and/or the LCDR1, LCDR2 and LCDR3 sequences of one of the light chain variable regions shown in SEQ ID NOs: 20, 22, 24, 26, 28, and 30, or a variant of the CDR sequence combination.
  • the present invention also provides an anti-ANG-2 antibody, or antigen-binding fragment thereof that binds to the epitope same as or overlapping with that bound by, and/or competes for binding to ANG-2 with, and/or inhibits (e.g., competitively inhibits) an exemplary antibody of the present invention, such as an antibody having a combination of VH and VL sequences as shown in Table B.
  • the present invention provides a nucleic acid encoding an antibody or antigen-binding fragment of the invention, a vector comprising the nucleic acid and a host cell comprising the vector.
  • the present invention provides a method of preparing an antibody or antigen-binding fragment of the invention.
  • the present invention provides an immunoconjugate, a multispecific antibody, a pharmaceutical composition, and a combination product comprising an antibody of the invention.
  • the present invention is also directed to a method of detecting a human and/or cynomolgus monkey ANG-2 in a sample.
  • the present invention also provides a method of blocking an ANG-2-mediated biological activity in a subject by using an antibody of the invention.
  • the present invention provides a method of preventing or treating a vascular-related disease, such as ocular neovascularization diseases, pulmonary inflammation, and various solid tumors, by using an antibody of the invention.
  • a vascular-related disease such as ocular neovascularization diseases, pulmonary inflammation, and various solid tumors
  • the present invention provides a use of an anti-ANG2 antibody of the invention in treatment of a disease or a disorder associated with vascular leakage and/or vascular inflammatory condition.
  • the diseases or disorders treatable by the methods of the invention include any diseases or disorders that may benefit from the blocking (i.e., ameliorating, inhibiting, relieving, or alleviating) of increased vascular endothelial permeability mediated by ANG2 and/or vascular inflammatory conditions associated with an ANG2 activity, such as any diseases or disorders accompanied with vascular leakage and/or vascular inflammation.
  • diseases or disorders include, but are not limited to, diabetic macular edema, retinal vein occlusion, wet age-related macular degeneration (wet AMD), diabetic retinopathy, cancer, sepsis, capillary leakage syndrome, pulmonary inflammatory condition, angioedema and vascular leakage syndrome.
  • the disease or disorder is a pulmonary inflammatory condition, such as pulmonary inflammation caused by infection with, for example, a virus or bacterial, e.g., viral pneumonia, and acute lung injury or acute respiratory distress syndrome caused, for example, by pulmonary inflammation.
  • the disease or disorder is acute lung injury (ALI).
  • the disease or condition is acute respiratory distress syndrome.
  • the present invention provides a method for reducing vascular permeability, comprising administering to a subject in need thereof a therapeutically effective amount of an anti-ANG2 antibody or antigen-binding fragment thereof of the invention or a pharmaceutical composition comprising the same.
  • the present invention provides a method of reducing vascular leakage and/or ameliorating a vascular inflammatory condition comprising administering to a subject in need thereof a therapeutically effective amount of an anti-ANG2 antibody or antigen-binding fragment thereof of the invention or a pharmaceutical composition comprising the same.
  • the present invention provided a method for preventing and/or treating a disease or disorder accompanied with vascular leakage, comprising administering to a subject in need thereof a therapeutically effective amount of an anti-ANG2 antibody or antigen-binding fragment thereof of the invention or a pharmaceutical composition comprising the same.
  • the present invention provides a method for preventing and/or treating a disease or disorder accompanied with vascular inflammation, comprising administering to a subject in need thereof a therapeutically effective amount of an anti-ANG2 antibody or antigen-binding fragment thereof of the invention or a pharmaceutical composition comprising the same.
  • the present invention provides a method for preventing and/or treating a pulmonary inflammatory condition, especially acute respiratory distress syndrome and acute lung injury, comprising administering to a subject in need thereof a therapeutically effective amount of an anti-ANG2 antibody or antigen-binding fragment thereof of the invention or a pharmaceutical composition comprising the same.
  • the present invention relates to a method of ameliorating one or more of the following disease parameters of acute respiratory distress syndrome: infiltration intensity of inflammatory cells in lung tissues, blood oxygen saturation, pulmonary edema degree, lung weight index, intensity of inflammatory lesions in lung tissues, lung CT images, and survival rate, comprising administering to a subject in need thereof a therapeutically effective amount of an anti-ANG2 antibody or antigen-binding fragment thereof of the invention or a pharmaceutical composition comprising the same.
  • acute respiratory distress syndrome is caused by pulmonary inflammatory injury, e.g., bacterial or viral infection or an inflammatory agent such as bacterial endotoxin LPS.
  • pulmonary inflammatory injury e.g., bacterial or viral infection or an inflammatory agent such as bacterial endotoxin LPS.
  • the method is a prophylactic for an individual at risk of developing ARDS.
  • the method is a therapeutic for an individual who has been diagnosed or suspected of having ARDS.
  • the anti-ANG2 antibody of the invention blocks the binding of human ANG-2 to human TIE2 expressed on the surface of vascular endothelial cells and/or blocks the binding of human ANG-2 to an integrin.
  • the anti-ANG2 antibody of the invention inhibites intracellular Tie2 phosphorylation induced by human ANG-2.
  • the anti-ANG2 antibody of the invention comprises an HCDR1 sequence of SEQ ID NO: 1, an HCDR2 sequence of SEQ ID NO: 2, an HCDR3 sequence of SEQ ID NO: 3, an LCDR1 sequence of SEQ ID NO: 4, an LCDR2 sequence of SEQ ID NO: 5, and an LCDR3 sequence of SEQ ID NO: 6. More preferably, the ANG2 antibody of the invention comprises a heavy chain variable region VH sequence of SEQ ID NO: 25 and a light chain variable region VL sequence of SEQ ID NO: 26.
  • the anti-ANG2 antibody of the invention is administered as a monotherapy agent. In some embodiments, the anti-ANG2 antibody of the invention is administered in combination with a second therapeutic agent. In yet some other embodiments, the second therapeutic agent is an anti-VEGF inhibitor molecule. In yet other embodiments, the anti-ANG2 antibody of the invention is administered independently of (i.e., not in combination with) a VEGF pathway-blocking agent, such as an anti-VEGF inhibitor.
  • the term “comprise” or “include” means the inclusion of a stated element(s), integer(s), or step(s), but not the excludion of any other element(s), integer(s), or step(s).
  • the term “comprise” or “include” encompasses the situations consisting of the stated element(s), integer(s) or step(s) unless otherwise indicated.
  • a variable region of an antibody that "comprises” a particular sequence it is also intended to encompass a variable region of an antibody that consists of that particular sequence.
  • antibody refers to a polypeptide comprising at least a light or heavy chain immunoglobulin variable region that specifically recognizes and binds to an antigen.
  • This term covers a variety of antibody structures, including, but not limited to, monoclonal antibodies, polyclonal antibodies, single- or multiple-chain antibodies, monospecific or multispecific antibodies (e.g., bispecific antibodies), chimeric or humanized antibodies, full-length antibodies, and antibody fragments, as long as they exhibit the desired antigen-binding activities.
  • a “whole antibody” (used interchangeably herein with “full-length antibody”, “complete antibody” and “intact antibody”) comprise at least two heavy chains (H) and two light chains (L).
  • Each heavy chain consists of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region consists of 3 domains, CH1, CH2, and CH3.
  • Each light chain consists of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the light chain constant region consists of one domain, CL.
  • Variable regions are domains in the heavy or light chains of antibodies that are involved in the binding of the antibodies to their antigen.
  • Constant regions are not directly involved in antibody-antigen binding, but they display a variety of effector functions.
  • the light chains of antibodies can be assigned to one of two types, known as kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequences of light chain constant domains.
  • the heavy chains of antibodies can be divided into five major types, IgA, IgD, IgE, IgG, and IgM, depending on the amino acid sequences of heavy chain constant regions, , and several of these types can be further divided into subtypes, such as IgG1, IgG2, IgG3, and IgG4, IgA1, and IgA2.
  • the heavy chain constant regions corresponding to different antibody types are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • isotype refers to the type of an antibody that is determined by the heavy chain constant region of the antibody. See generally, Fundamental Immunology, Ch.7 (Paul, W. Ed., 2nd ed., Raven Press, N.Y. (1989 )) (which is incorporated herein by reference in its entirety and for all purposes).
  • a full-length antibody comprising two heavy chains and two light chains, especially an antibody of IgG type, is used in the methods of the present invention.
  • antigen-binding portion of an antibody refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds to the antigen to which the intact antibody binds.
  • the antigen-binding portion of an antibody typically comprises amino acid residues from a "complementary determining region" or a "CDR”.
  • An antigen-binding fragment can be prepared using recombinant DNA technology, or by enzymatic or chemical cleavage of an intact antibody.
  • Antigen-binding fragments include, but are not limited to, Fab, scFab, Fab', F (ab') 2 , Fab'-SH, Fv, single-chain Fv, diabody, triabody, tetrabody, minibody, and single-domain antibody (sdAb). More detailed description of antibody fragments can be found in Fundamental Immunology, edited by W. E. Paul, Raven Press, N.Y. (1993 ); Shao Rongguang, et al. (Editor), Research and Application of Antibody-based Drugs. People's Medical Publishing House (2013 ); Hollinger et al., PNAS USA 90:6444-6448(1993 ); and Hudson et al., Nat. Med. 9: 129-134 (2003 ).
  • antibodies or antigen-binding fragments of the present invention may comprise the following configurations of variable and constant regions: (i) VH-CH1; (ii) VH-CH2; (iii) VH-CH3; (iv) VH-CH1-CH2; (v) VH-CH1-CH2-CH3; (vi) VH-CH2-CH3; (vii) VH-CL; (viii) VL-CH1; (ix) VL-CH2; (x) VL-CH3; (xi) VL-CH1-CH2; (xii) VL-CH1-CH2-CH3; (xiii) VL-CH2-CH3; and (xiv) VL-CL.
  • chimeric antibody refers to an antibody in which the variable region sequence is derived from one species and the constant region sequence is derived from another species, e.g., an antibody in which the variable region sequence is derived from a mouse antibody and the constant region sequence from a human antibody.
  • humanized antibody refers to an antibody in which CDR sequences derived from another mammalian species (such as mice) are grafted into the framework region sequences of human. Additional modifications to framework regions can be made within the human framework region sequences.
  • an “isolated” antibody is one that has been separated from a component of its natural environment.
  • the antibody is purified to greater than 95% or 99% purity, as determined by, for example, electrophoresis (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatography (e.g., ion exchange or reverse phase HPLC).
  • electrophoresis e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatography e.g., ion exchange or reverse phase HPLC
  • epitope refers to a region in an antigen to which an antibody binds. Epitopes can be formed by contiguous amino acids or non-contiguous amino acids juxtaposed by tertiary folding of a protein.
  • ANG-2 and “ANG2” are used interchangeably herein and refer to angiopoietin 2.
  • the expressions include variants, isoforms, homologs, and species homologs of ANG-2.
  • human ANG-2 refers to ANG-2 of a human sequence.
  • a specific human ANG-2 sequence is shown in SEQ ID NO: 51.
  • a human ANG-2 sequence has at least 95%, or even 96%, 97%, 98%, or 99% amino acid sequence identity to the human ANG-2 amino acid sequence as shown in SEQ ID NO: 51.
  • Amino acid sequences of ANG-2 proteins of other species, such as mouse and monkey ANG-2 sequences are available from the NCBI protein sequence database, e.g.
  • ANG-2 encompasses fragments of ANG-2, such as fragments capable of exerting a biological function of a full-length ANG-2 molecule, e.g., fragments comprising a C-terminal fibrinogen-like domain (FLD).
  • ANG2 if not explicitly indicated as from a non-human species (e.g., "murine ANG2" or “monkey ANG2”), refers to ANG2 from human, or a biologically active fragment thereof.
  • ANG1 refers to angiopoietin 1.
  • the antibodies of the present invention bind to ANG2, but not to ANG1, although ANG2 and ANG1 share a considerable amino acid sequence homology and competitively bind to the same site of the TIE2 receptor.
  • An amino acid sequence of human ANG1 is available from the NCBI Protein Sequence Database under accession number AAB50557.
  • the term ANG1, if not explicitly indicated as from a non-human species e.g., "murine ANG1" or “monkey ANG1”
  • TIE2 refers to tyrosine kinase 2 with Ig and EGF homology domains.
  • TIE2 refers to TIE2 from human or a biologically active fragment thereof.
  • An amino acid sequence of human TIE2 is available from the NCBI Protein Sequence Database under accession number AAA61130.
  • the term "specifically bind” means that an antibody selectively or preferentially binds to an antigen. If an antibody binds to human ANG-2 with a K D of about 5 ⁇ 10 -7 M or less, about 1 ⁇ 10 -7 M or less, about 5 ⁇ 10 -8 M or less, about 1 ⁇ 10 -8 M or less, or about 5 ⁇ 10 -9 M or less in a biological optical interferometry assay, the antibody is considered as an antibody that "specifically binds to human ANG-2". However, antibodies that specifically bind to human ANG-2 can be cross-reactive with ANG-2 proteins from other species.
  • an antibody specific for human ANG-2 is cross-reactive with ANG-2 protein from cynomolgus monkeys, but not with murine ANG-2.
  • Methods of determining cross-reactivity include the methods described herein in the Examples and the standard assays known in the art, for example, by using biological optical interferometry or flow cytometry.
  • affinity refers to the intrinsic binding affinity that reflects the interactions between members of a binding pair.
  • the affinity of a molecule X for its partner Y can generally be represented by an equilibrium dissociation constant (K D ), which is the ratio of a dissociation rate constant to an association rate constant (k dis and k on , respectively).
  • K D equilibrium dissociation constant
  • k dis and k on association rate constant
  • Affinity can be measured by common methods known in the art.
  • a specific method for measuring affinity is the ForteBio kinetic binding assay described herein.
  • high affinity for an IgG antibody means that the antibody binds to a target antigen with a K D of 1 ⁇ 10 -7 M or less, preferably 5 ⁇ 10 -8 M or less, more preferably about 1 ⁇ 10 -8 M or less, and even more preferably about 5 ⁇ 10 -9 M or less.
  • a "high affinity” for binding may vary with different antibody isotypes.
  • "high affinity” means that the antibody has a K D of 1 ⁇ 10 -6 M or less, preferably 1 ⁇ 10 -7 M or less, more preferably about 1 ⁇ 10 -8 M or less.
  • antibodies of the present invention are ANG2 neutralizing antibodies or blocking antibodies that, upon binding to ANG2, block the interaction of ANG2 with its receptor (TIE2) and/or with an integrin and/or result in inhibition of at least one biological activity of ANG2.
  • TIE2 its receptor
  • Detection of the activity of the neutralizing antibodies and the blocking antibodies can be performed using the assays known in the art or described in this disclosure, such as those exemplified in the Examples.
  • an " antibody that competes for binding" with a reference antibody that binds to an antigen refers to an antibody that blocks the binding of the reference antibody to the antigen (e.g., ANG-2) by 50% or more in a competition assay, and conversely, the reference antibody blocks the binding of the antibody to the antigen (e.g., ANG-2) by 50% or more in a competition assay.
  • exemplary competition assays are described in: "Antibodies", Harlow and Lane (Cold Spring Harbor Press, Cold Spring Harbor, NY ).
  • the antibodies that compete for binding may bind to the same epitope region as the reference antibody, e.g., the same epitopes, adjacent epitopes, or overlapping epitopes.
  • An antibody that inhibits (e.g., competitively inhibits) the binding of a reference antibody to its antigen refers to an antibody that inhibits the binding of the reference antibody to its antigen by more than 50%, 60%, 70%, 80%, 90% or 95%. Conversely, the reference antibody also inhibits the binding of the antibody to its antigen by more than 50%, 60%, 70%, 80%, 90%, or 95%.
  • the binding of an antibody to its antigen can be measured by affinity, such as an equilibrium dissociation constant. Methods for affinity determination are known in the art.
  • An antibody that exhibits the same or similar binding affinity and/or specificity as a reference antibody refers to an antibody that is capable of exhibiting at least 50%, 60%, 70%, 80%, 90%, or more than 95% of the binding affinity and/or specificity of the reference antibody. This can be determined by any method known in the art for determining the binding affinity and/or specificity.
  • Fc region is used herein to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native sequence Fc-regions and variant Fc-regions.
  • a human IgG heavy chain Fc region extends from Cys226 or from Pro230 to the carboxy terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc-region may or may not be present.
  • the numbering of amino acid residues in the Fc-region or constant region is according to the EU numbering system, also known as the EU Index, as described in Kabat, E. A. et al., Sequences of Proteins of Immunological Interest, 5th edition, Public Health Service, National Institutes of Health, Bethesda, MD (1991), NIH Publication 91-3242 .
  • variant with reference to an antibody, is used herein to refer to an antibody comprising an antibody region of interest that has been modified in amino acids by substitution, deletion and/or insertion of at least one amino acids, e.g., 1-30 amino acids, or 1-20 amino acids or 1-10 amino acids (e.g., 1, 2, 3, 4 or 5 amino acids), as compared to a reference antibody, wherein the variant substantially retains at least one biological property (e.g., antigen-binding capacity) of the original antibody molecule without the modification.
  • the antibody region of interest may be the full length of the antibody, or the heavy chain variable region, the light chain variable region, or the combination thereof, or the heavy chain CDR region(s) or the light chain CDR region(s) or the combination thereof.
  • an antibody region that has amino acid modifications is also referred to as a "variant" of that antibody region.
  • sequence identity refers to the degree to which a sequence is identical to another on a nucleotide-by-nucleotide or amino acid-by-amino acid basis in a comparison window.
  • the “percentage of sequence identity” can be calculated using the following method: comparing two optimally aligned sequences in a comparison window to determine the number of the positions where the same nucleic acid base (e.g., A, T, C, G, I) or the same amino acid residue (e.g., Ala, Pro, Ser, Thr, Gly, Val, Leu, Ile, Phe, Tyr, Trp, Lys, Arg, His, Asp, Glu, Asn, Gln, Cys, and Met) is present in the two sequences so as to obtain the number of matching positions, then dividing the number of matching positions by the total number of the positions in the comparison window (i.e., window size), and multiplying the result by 100 to obtain the percentage of sequence identity.
  • nucleic acid base e.g
  • the optimal alignment for determining the percent sequence identity can be achieved in a variety of ways known in the art, for example, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DASTAR) software.
  • BLAST BLAST-2
  • ALIGN ALIGN
  • DASTAR Megalign
  • One skilled in the art can determine suitable parameters for aligning sequences, including any algorithm required to achieve maximum alignment over the full length of the sequence being compared or within a sequence region of interest.
  • the percentage of amino acid sequence identity is determined by optimally aligning a candidate antibody sequence with a reference antibody sequence, and in a preferred embodiment by optimally aligning according to the Kabat numberings. In the present disclosure, if not specifying a comparison window (i.e., the antibody region of interest to be compared), the alignment is made over the full length of the reference antibody sequence .
  • sequence identity may be present along the entire heavy chain variable regions and/or the entire light chain variable regions of antibodies, or sequence percentage identity may be limited to the framework regions only, with the sequences in corresponding CDR regions remaining 100% identical.
  • a candidate antibody with an amino acid modification in an antibody region of interest, as relative to a reference antibody can be identified based on sequence alignment.
  • a "conservative substitution” means an amino acid modification by which an amino acid is replaced with a chemically similar amino acid.
  • Amino acid modifications such as substitutions can be introduced into the antibodies of the present invention using standard methods known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis.
  • a table of conservative substitutions providing functionally similar amino acids is well-known in the art.
  • residues for conservative substitution are from the following conservative substitution table, Table A, and preferably are the residues under preferred conservative substitutions in Table A.
  • the antibodies of the present invention used in the methods of the present invention are antibody sequences having conservative substitutions, for example, antibody sequences that are derived from an exemplary antibody sequence of the invention (e.g., any one or combination of SEQ ID Nos: 1 to 10) by introducing one or more conservative substitutions, especially preferred conservative substitutions in Table A.
  • mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • domesticated animals e.g., cows, sheep, cats, dogs, and horses
  • primates e.g., humans and non-human primates such as monkeys
  • rabbits e.g., mice and rats
  • rodents e.g., mice and rats
  • treatment includes 1) a therapeutic measure that cures, postpones, alleviates the symptoms of and/or halts the progression of a diagnosed pathological condition or a disorder; and 2) a prophylactic or preventive measure that prevents and/or postpones the development of a pathological condition or a disorder.
  • subjects in need of treatment include individuals having already developed a disorder, individuals susceptible to a disorder, and individuals to be prevented from a disorder. The individuals will benefit from the therapeutic or prophylactic measure and exhibit a reduction or improvement in the occurrence, recurrence, or progression of a disease, disorder, condition, and/or symptom as compared to an individual not receiving the said treatment.
  • the present invention involves the treatment of a disease or a disorder. In other embodiments, the present invention involves the prevention of a disease or a disorder.
  • treatment of a disease or a disorder means amelioration of the disease or the disorder (i.e., slowing or preventing or reducing the progression or at least one clinical symptom). In some other embodiments, “treatment” means relief or improvement of at least one physical parameter, including those physiological parameters that may be indiscernible by patients. In some other embodiments, “treatment” means modulating a disease or a disorder physically (e.g., stabilizing a discernible symptom), physiologically (e.g., stabilizing a physical parameter), or both. Unless explicitly otherwise indicated herein, methods for assessing the therapeutic and/or prophylactic efficacy of a disease or disorder are generally known in the art.
  • treatment of a disease or a disorder means “prevention” of a disease or a disorder, including inhibition of the occurrence or progression of the disease or the disorder or a particular symptom of the disease or the disorder.
  • the subject is a candidate for a prevention regimen.
  • prevention refers to administration of a drug prior to the appearance of any signs or symptoms of a disease or a disorder, particularly in a subject at risk of developing a disease.
  • therapeutically effective amount refers to an amount of an anti-ANG2 antibody or antigen-binding fragment thereof of the present invention, which, when administered to a cell, tissue or subject alone or in combination with additional therapeutic agent, is effective in preventing or ameliorating one or more symptoms of or the progression of a disease or a disorder.
  • a therapeutically effective amount also refers to an amount of an antibody or antigen-binding fragment thereof that is sufficient to result in the improvement of a symptom, such as an amount that results in treatment, remedy, prevention, or amelioration of a relevant medical condition or an increase in the rate of treatment, remedy, prevention, or amelioration of such a condition.
  • the therapeutically effective amount refers to the amount of the agent alone.
  • the therapeutically effective amount refers to the combined amount of the active agents that cause a therapeutic effect, whether the active agents are administered concomitantly, sequentially, or simultaneously.
  • An effective amount of a therapeutic agent will result in an improvement in a diagnostic criteria or parameter of at least 10%, typically at least 20%, preferably at least about 30%, more preferably at least 40%, and most preferably at least 50%.
  • a VEGF blocker refers to an anti-VEGF agent that blocks a function/activity of VEGF or VEGF receptor.
  • Anti-VEGF agents include, for example, monoclonal antibodies specifically binding to VEGF-A, which agents bind to the VEGF to block the binding of the VEGF to its receptor; and VEGFR blocking agents consisting of an extracellular fragment of a VEGF receptor and an Fc region of an immunoglobulin, which agents have an inherently high specificity for VEGF.
  • anti-VEGF agents include VEGF-Trap (see, e.g., US7,087,411 ), anti-VEGF antibodies (e.g., bevacizumab), small molecule kinase inhibitors for VEGF receptors (e.g., sunitinib).
  • a second therapeutic agent for use in combination with an antibody of the present invention is a VEGF blocker.
  • an antibody of the invention is administered independently of (i.e., not in combination with) a VEGF blocker.
  • the present invention provides antibodies or antigen-binding fragments thereof that specifically bind to ANG-2, preferably human ANG-2 protein (e.g., a human ANG-2 sequence of SEQ ID NO: 51).
  • the antigen-binding fragments of the antibodies of the present invention are an antibody fragment selected from the following antibody fragments: Fab, Fab', Fab'-SH, Fv, a single-chain antibody such as scFv, (Fab') 2 fragment, a single-domain antibody, a diabody (dAb), and a linear antibody.
  • the antibodies of the invention have one or more advantageous properties selected from the group consisting of:
  • the antibodies or antigen-binding fragments thereof of the present invention exhibit at least one, more preferably at least two, more preferably at least three, four, or five of the above properties, and even more preferably all of the above properties.
  • the antibodies of the invention are a humanized antibody.
  • the antibodies of the invention are antibodies produced and isolated from mammalian producer cells, such as CHO cells.
  • Complementarity determining region or “CDR region” or “CDR” (used interchangeably herein with hypervariable region “HVR”) is an amino acid region in the variable region of an antibody that is predominantly responsible for binding to an epitope of an antigen.
  • the CDRs of heavy and light chains are commonly referred to as CDR1, CDR2, and CDR3, numbered sequentially from the N-terminus.
  • the CDRs located within the variable domain of an antibody heavy chain are referred to as HCDR1, HCDR2, and HCDR3, while the CDRs located within the variable domain of an antibody light chain are referred to as LCDR1, LCDR2, and LCDR3.
  • VH and VL sequence combinations of some exemplary antibodies of the invention are given in Table B below:
  • Antibody VH comprising or consisting of an amino acid sequence shown in the following SEQ ID NO VL comprising or consisting of an amino acid sequence shown in the following SEQ ID NO 1 SEQ ID NO:19 SEQ ID NO:20 2 SEQ ID NO:21 SEQ ID NO:22 3 SEQ ID NO:23 SEQ ID NO:24 4 SEQ ID NO:25 SEQ ID NO:26 5 SEQ ID NO:27 SEQ ID NO:28 6 SEQ ID NO:29 SEQ ID NO:30
  • an antibody of the invention comprises: (i) the HCDR1, HCDR2 and HCDR3 sequences contained in the VH of SEQ ID NO: 25; and (ii) the LCDR1, LCDR2 and LCDR3 sequences contained in the VL of SEQ ID NO: 26.
  • CDRs Kabat complementarity determining regions
  • Chothia scheme refers to the positions of the structural loops ( Chothia and Lesk, J. Mol. Biol. 196: 901-917 (1987 )).
  • AbM HVRs are a compromise between Kabat HVRs and Chothia structural loops and are used by Oxford Molecular's AbM antibody modeling software.
  • Contact HVRs are based on analysis of available complex crystal structures. Residues of each HVR/CDR of these HVRs according to the different CDR determination shemes are described below.
  • CDR Kabat scheme AbM scheme Chothia scheme Contact scheme LCDR1 L24-L34 L24-L34 L26-L32 L30-L36 LCDR2 L50-L56 L50-L56 L50-L52 L46-L55 LCDR3 L89-L97 L89-L97 L91-L96 L89-L96 HCDR1 H31-H35B H26-H35B H26-H32 H30-H35B (Kabat numbering system) HCDR1 H31-H35 H26-H35 H26-H32 H30-H35 (Chothia numbering system) HCDR2 H50-H65 H50-H58 H53-H55 H47-H58 HCDR3 H95-H102 H95-H102 H96-H101 H93-H101 (Kabat numbering system)
  • HVRs can also be HVR sequences located according to the Kabat numbering system at the following Kabat residue positions: Positions 24-36 or 24-34 (LCDR1), positions 46-56 or 50-56 (LCDR2), and positions 89-97 or 89-96 (LCDR3) in VL; and positions 26-35 or 27-35B (HCDR1), positions 50-65 or 49-65 (HCDR2), and positions 93-102, 94-102, or 95-102 (HCDR3) in VH.
  • LCDR1 Positions 24-36 or 24-34
  • LCDR2 positions 46-56 or 50-56
  • LCDR3 positions 89-97 or 89-96
  • the HVRs of an antibody of the present invention are HVR sequences located according to the Kabat numbering system at the following Kabat residue positions:
  • HVRs can also be determined based on the same positions according to Kabat numbering as a reference CDR sequence (e.g., any one of the exemplary CDRs of the invention, for example, the sequences of SEQ ID NOs: 1-6).
  • CDR or “CDR sequence” or “HVR” or “HVR sequence” used herein includes HVR or CDR sequences determined in any of the ways described above.
  • residue positions in an antibody variable region are numbered according to the Kabat numbering system ( Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991 )).
  • Combination HCDR1 comprising or consisting of the amino acid sequence shown in the following SEQ ID NO HCDR2, comprising or consisting of the amino acid sequence shown in the following SEQ ID NO HCDR3, comprising or consisting of the amino acid sequence shown in the following SEQ ID NO LCDR1, comprising or consisting of the amino acid sequence shown in the following SEQ ID NO LCDR2, comprising or consisting of the amino acid sequence shown in the following SEQ ID NO LCDR3, comprising or consisting of the amino acid sequence shown in the following SEQ ID NO 1 SEQ ID NO:1 SEQ ID NO:2 SEQ ID NOT SEQ ID NO:4 SEQ ID NO:5 SEQ ID NO:6 2 SEQ ID NO:7 SEQ ID NO:8 SEQ ID NO:9 SEQ ID NO:10 SEQ ID NO:11 SEQ ID NO:12 3 SEQ ID NO:13 SEQ ID NO:14 SEQ ID NO:15 SEQ ID NO:16 SEQ ID NO:17 SEQ ID NO:
  • an antibody or antigen-binding fragment thereof of the invention comprises a heavy chain variable region and a light chain variable region, wherein the antibody comprises:
  • an antibody or antigen-binding fragment thereof of the invention comprises:
  • an antibody or antigen-binding fragment thereof of the invention comprises three complementarity-determining regions of a heavy chain variable region (HCDRs), and three complementarity-determining regions of a light chain variable region (LCDRs), wherein,
  • an antibody or antigen-binding fragment thereof of the invention comprises three complementarity-determining regions of a heavy chain variable region (HCDRs), and three complementarity-determining regions of a light chain variable region (LCDRs), wherein,
  • an antibody or antigen-binding fragment thereof of the invention comprises three complementarity-determining regions of a heavy chain variable region (HCDRs), and three complementarity-determining regions of a light chain variable region (LCDRs), wherein,
  • the present invention also contemplates antibodies comprising variants of the CDR sequences disclosed herein.
  • the smallest overlapping region can be determined to provide a "minimum binding unit" for antigen binding.
  • the residues in the remaining portions of the CDR sequences outside the minimum binding unit can be determined by the structure and protein folding of the antibody.
  • the amino acid residues of the minimal binding unit may remain unchanged, while one or more of the remaining CDR residues according to the Kabat or Chothia scheme are conservatively substituted, such as by those conservative substitution residues presented in Table A.
  • each CDR may be modified individually or in combination.
  • the amino acid modification is an amino acid substitution, especially a conservative amino acid substitution, such as a preferred conservative amino acid substitution set out in Table A.
  • the amino acid substitution occurs at an amino acid position corresponding to an X residue of a consensus CDR sequence provided herein (e.g., SEQ ID NO: 54, 55, 56, 57, 58, or 59).
  • a “variable region” or “variable domain” is a domain in the heavy or light chain of an antibody that is involved in binding of the antibody to antigen thereof.
  • a heavy chain variable region (VH) and a light chain variable (VL) region can be further subdivided into hypervariable regions (HVRs, also known as complementarity-determining regions (CDRs)), interspersed with more conservative regions (i.e., framework regions (FRs)).
  • HVRs hypervariable regions
  • CDRs complementarity-determining regions
  • FRs framework regions
  • Each VH or VL consists of three CDRs and four FRs, arranged in the following order from amino-terminus to carboxy-terminus: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • one or more residues in one or both of the two variable regions (i.e., VH and/or VL) of an antibody can be modified, for example, a residue modification, especially a conservative residue substitution, can be made in one or more CDR regions and/or in one or more framework regions, while the resulted modified antibody still substantially retains at least one biological property (e.g., ability to bind to antigen) as compared to before the modification.
  • the binding properties or other functional properties of a mutated antibody can be assessed by in vitro or in vivo assays.
  • a framework region residue can be mutated, e.g., to improve a property of an antibody.
  • one or more framework residues can be " back-mutated" to the residue(s) of a corresponding germline sequence.
  • CDR grafting is another type for antibody variable region modification known in the art. Since CDR sequences are responsible for most antibody-antigen interactions, a recombinant antibody variant can be constructed that mimic the properties of a known antibody. In such an antibody variant, CDR sequences from a known antibody are grafted onto framework regions from a different antibody with a different property.
  • the present invention relates to an anti-ANG-2 antibody, or antigen-binding fragment thereof, comprising CDR sequences from the heavy and light chain variable regions of one of the antibodies presented in Table B, but with different framework region sequences.
  • the framework region sequences for such substitution can be obtained from public DNA databases, including germline antibody gene sequences, or from published anti-ANG-2 antibody sequences.
  • germline DNAs for human heavy and light chain variable region genes are available from the GenBank database.
  • the sequence of the antibody protein can be compared with the protein sequences in databases using sequence similarity search tools, such as Gapped BLAST.
  • sequence similarity search tools such as Gapped BLAST.
  • the framework sequence for substitution has a structural similarity, e.g., a sequence identity of at least 80%, 85% or 90%, or more than 95%, 96%, 97%, 98% or 99%.
  • an antibody of the invention comprises, or consists of, the heavy chain variable region VH sequence of any of the antibodies listed in Table B.
  • an antibody of the invention comprises a variant of said VH sequence.
  • an antibody of the invention comprises, or consists of, the light chain variable region VL sequence of any of the antibodies listed in Table B. In yet another embodiment, an antibody of the invention comprises a variant of said VL sequence.
  • an antibody of the invention comprises:
  • the variant of the VH sequence has at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or greater identity in amino acid sequence compared to the reference VH sequence, preferably over the full length or within the three regions of CDR1, 2 and 3.
  • the variant of the VH sequence comprises at least one and no more than 30, 10, or 5, 4, 3, 2, 1, 0 amino acid modifications (preferably amino acid substitutions, preferably conservative substitutions) in amino acid sequence compared to the reference VH sequence, preferably, over the full length or within the three regions of CDR1, 2, and 3.
  • any of the sequence differences do not occur in the CDR regions.
  • the variant of the VL sequence has at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or greater identity in amino acid sequence compared to the reference VL sequence, preferably over the full length or within the three regions of CDR1, 2 and 3.
  • the variant of the VL sequence comprises at least one and no more than 30, 10, or 5, 4, 3, 2, 1, 0 amino acid modifications (preferably amino acid substitutions, preferably conservative substitutions) in amino acid sequence compared to the reference VL sequence, preferably over the full length or within the three regions of CDR1, 2, and 3.
  • any of the sequence differences do not occur in the CDR regions.
  • an antibody of the invention comprises or consists of the VH/VL sequence pairs of the heavy chain variable region and the light chain variable region of any of the antibodies presented in Table B.
  • the present invention also provides variants of the antibody, such as variants having at least 95% to 99% identity or comprising no more than 10 amino acid modifications in the VH, the VL, or both the VH and the VL. Preferably any of the sequence differences do not occur in the CDR regions.
  • the present invention provides an antibody, or antigen-binding fragment thereof, comprising:
  • the present invention provides an antibody, or antigen-binding fragment thereof, comprising a heavy chain variable region and a light chain variable region selected from the group consisting of:
  • an antibody of the invention comprises a heavy chain variable region, wherein the heavy chain variable region comprises or consists of the sequence of SEQ ID NO: 25.
  • an antibody of the invention comprises a light chain variable region, wherein the light chain variable region comprises or consists of the sequence of SEQ ID NO: 26.
  • an antibody of the present invention comprises a heavy chain variable region with VH sequence of SEQ ID NO: 25 and a light chain variable region with VL sequence of SEQ ID NO: 26.
  • an antibody of the invention comprises:
  • the present invention relates to anti-ANG-2 antibodies, or antigen-binding fragments thereof, and their uses, wherein the antibodies comprise the CDR sequences contained in the heavy chain variable region of SEQ ID NO: 25 and the light chain variable region of SEQ ID NO: 26, but having different framework region sequences.
  • the framework sequences for substitution have structural similarity, e.g., a sequence identity of at least 80%, 85% or 90%, or more than 95%, 96%, 97%, 98% or 99%.
  • Antibody heavy chains and light chains are Antibody heavy chains and light chains
  • an antibody of the invention comprises a heavy chain Fc region, such as an Fc region of an IgG1, IgG2 or IgG4 isotype. In one embodiment, an antibody of the invention comprises an IgG1 Fc region. In some embodiments, an antibody of the invention comprises a kappa light chain constant region, such as a human kappa light chain constant region.
  • the Fc region comprises the amino acid sequence of SEQ ID NO: 52, or an amino acid sequence comprising at least one, two or three, but not more than 20, 10 or 5 amino acid modifications relative to the amino acid sequence of SEQ ID NO: 52, or a sequence having at least 95% to 99% identity to the amino acid sequence of SEQ ID NO: 52.
  • an antibody of the invention comprises a light chain constant region.
  • the light chain constant region is a human kappa light chain constant region.
  • the light chain constant region comprises the amino acid sequence of SEQ ID NO: 53, or an amino acid sequence comprising at least one, two or three, but not more than 20, 10 or 5 amino acid modifications relative to the amino acid sequence of SEQ ID NO: 53, or a sequence having at least 95% to 99% identity to the amino acid sequence of SEQ ID NO: 53.
  • an antibody of the invention comprises a heavy chain, which comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 31, 33, 35, 37, 39, and 41, or an amino acid sequence comprising at least 1, 2 or 3, but no more than 20, 10 or 5 amino acid modifications relative thereto, or an amino acid sequence having at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or greater identity thereto.
  • the amino acid modifications do not occur in the CDR regions and, more preferably, do not occur in the variable regions.
  • an antibody of the invention comprises a light chain, which comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 32, 34, 36, 38, 40, and 42, or an amino acid sequence comprising at least 1, 2 or 3, but not more than 20, 10 or 5 amino acid modifications relative thereto, or an amino acid sequence having at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or greater identity thereto.
  • the amino acid modifications do not occur in the CDR regions and, more preferably, do not occur in the variable regions.
  • an antibody of the invention comprises a heavy chain sequence and/or a light chain sequence selected from:
  • a residue modification is introduced into a constant region of the antibody, for example, to alter a property of the antibody, such as an effector function.
  • an antibody of the present invention comprises a heavy chain sequence as shown in SEQ ID NO: 37 and a light chain sequence as shown in SEQ ID NO: 38.
  • the present invention provides antibodies that specifically bind to ANG-2 (e.g., human ANG-2), which were isolated and characterized as described in the Examples.
  • Figure 26 shows the VH and VL sequences in the variable regions of the exemplary antibodies of the present invention.
  • Figure 25 shows the exemplary CDR sequences of the antibodies.
  • the heavy and light chain amino acid sequences and the nucleotide sequences encoding the variable regions VH and VL of the exemplary antibodies of the invention are shown in the Sequence Listing.
  • the present invention provides variants of any of the antibodies described herein, especially the exemplary antibodies listed in Table B.
  • the antibody variant retains at least 60%, 70%, 80%, 90%, or 100% of a biological activity (e.g., antigen-binding activity) of the unmodified antibody.
  • the modification in the antibody variant do not cause loss of the antigen-binding activity, but optionally conferring a property such as an improved antigen-binding affinity and a different effector function.
  • an antibody of the invention has at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or greater sequence identity in heavy chain variable region, as compared to a reference antibody (e.g., one of the antibodies listed in Table D).
  • a reference antibody e.g., one of the antibodies listed in Table D.
  • an antibody of the invention has at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or greater sequence identity in light chain variable region, as compared to a reference antibody (e.g., one of the antibodies listed in Table D).
  • a reference antibody e.g., one of the antibodies listed in Table D.
  • an antibody of the invention has at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or greater sequence identity in heavy chain variable region and/or light chain variable region, as compared to a reference antibody (e.g., one of the antibodies listed in Table D).
  • the Fc region of an antibody can be altered.
  • the modification in the Fc region can be made alone or in combination with the modification to the framework and/or CDR regions described above.
  • the Fc region may be modified, for example, so as to alter one or more of the functions of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or antigen-dependent cytotoxicity.
  • an antibody of the invention can also be chemically modified (e.g., conjugated with PEG), or have altered glycosylation pattern.
  • the Fc region may comprise an Fc region having one or more amino acid substitutions that increase ADCC activity, e.g., substitutions at Fc region positions 298, 333, and/or 334 (EU numbering scheme for residues).
  • the Fc region can also be modified to result in altered (i.e., increased or decreased) C1q binding and/or complement-dependent cytotoxicity (CDC) (see, for example, US6,194,551 , WO99/51642 and Idusogie, E. E. et al., J. Immunol. 164 (2000): 4178-4184 ).
  • CDC complement-dependent cytotoxicity
  • the Fc region can be modified to increase or decrease its glycosylation degree and/or to alter its glycosylation pattern. Addition or deletion of glycosylation sites of the Fc region can be conveniently achieved by altering the amino acid sequence so as to create or remove one or more glycosylation sites. For example, one or more amino acid substitutions can be made to eliminate one or more glycosylation sites, thereby eliminating glycosylation at the sites.
  • Antibodies with altered glycosylation patterns can be prepared, such as low- or non-fucosylated antibodies with reduced contents of fucosyl residues or antibodies with increased bisecting GlcNac structures. Such altered glycosylation patterns have been shown to increase ADCC activities of antibodies.
  • the present invention also contemplates antibody variants having at least one galactose residue in an oligosaccharide attached to Fc region. These antibody variants may have an improved CDC function.
  • the present invention also contemplates antibody variants that have some but not all effector functions, which makes them ideal candidates for certain applications where the in vivo half-life of an antibody is important, but some effector functions, such as complement and ADCC, are unnecessary or deleterious.
  • Fc region may comprise a mutation that eliminates or attenuates an effector function, such as a human IgG1 Fc region harboring the mutations P329G and/or L234A and L235A, or a human IgG4 Fc region harboring the mutations P329G and/or S228P and L235E.
  • mutations that may facilitate antibody production in the cross-mAb format may be introduced into the Fc region, such as knob-into-hole mutations. See , e.g., WO2016075035 .
  • cysteine-engineered antibodies such as " thioMabs", in which one or more antibody residues are substituted with a cysteine residue.
  • the number of cysteine residues in the hinge region of an antibody can be altered, e.g., to facilitate the assembly of light and heavy chains or increase or decrease the stability of the antibody. See, e.g., U.S. Patent No. 5,677,425 .
  • the antibodies provided herein can be further modified to contain a non-protein moiety.
  • the moieties suitable for antibody derivatization include, but are not limited to, water soluble polymers.
  • Non-limiting examples of water-soluble polymers include, but not limited to, polyethylene glycol (PEG), which can be used, for example, to extend the half-life (e.g., in serum) of an antibody.
  • PEG polyethylene glycol
  • Methods for PEGylation of proteins are known in the art and can be applied to the antibodies of the invention. See, e.g., EP 0154 316 and EP 0401384 .
  • the present invention provides a nucleic acid that encodes any of the above anti-ANG-2 antibodies or fragments thereof.
  • a vector comprising the nucleic acid is also provided.
  • the vector is an expression vector.
  • a host cell comprising the nucleic acid or the vector is also provided.
  • the host cell is eukaryotic.
  • the host cell is selected from a yeast cell and a mammalian cell (e.g., a CHO cell or a 293 cell).
  • the host cell is prokaryotic.
  • the present invention provides a nucleic acid that encodes any of the above anti-ANG-2 antibodies or fragments thereof.
  • the nucleic acid may comprise a nucleic acid that encodes the amino acid sequence of the light chain variable region and/or the heavy chain variable region of the antibody, or comprise a nucleic acid that encodes the amino acid sequence of the light chain and/or the heavy chain of the antibody.
  • the polypeptides encoded by the polynucleotides exhibit ANG-2 antigen-binding ability when expressed in suitable expression vectors.
  • polynucleotide encoding at least one CDR region, and generally all three CDR regions, of the heavy chain VH or light chain VL of an ANG-2-binding antibody described herein above.
  • the polynucleotide encodes all or substantially all of the variable region sequence of the heavy chain and/or the light chain of the ANG2-binding antibody described herein above .
  • each antibody or polypeptide amino acid sequence may be encoded by a number of nucleic acid sequences due to codon degeneracy.
  • polynucleotide sequences can be generated by de novo solid-phase DNA synthesis or by PCR mutagenesis of the sequences encoding antibodies or antigen-binding fragments thereof that bind ANG-2 using methods well known in the art.
  • a vector(s) comprising a nucleic acid of the invention is provided.
  • the vector is an expression vector, such as an eukaryotic expression vector.
  • Vectors include, but are not limited to, viruses, plasmids, cosmids, lambda phages, or yeast artificial chromosomes (YAC).
  • YAC yeast artificial chromosomes
  • the expression vector of the invention is a pTT5 expression vector.
  • a host cell comprising the vector.
  • Host cells suitable for cloning or expressing vectors encoding antibodies include prokaryotic or eukaryotic cells described herein.
  • antibodies can be produced in bacteria, particularly when glycosylation and Fc effector function are not required.
  • antibodies or antibody fragments can be expressed in E. coli. After expression, the antibodies can be isolated from bacterial cell pastes or soluble fractions, and further purified.
  • the host cell is eukaryotic.
  • the host cell is selected from yeast cells, mammalian cells, or other cells suitable for producing an antibody or antigen-binding fragment thereof.
  • eukaryotic microorganisms such as filamentous fungi or yeasts are suitable hosts for cloning or expressing vectors that encode antibodies.
  • fungal and yeast strains in which the glycosylation pathway has been "humanized” can generate antibodies with partially or fully humanized glycosylation patterns.
  • Host cells suitable for expressing glycosylated antibodies can also be derived from multicellular organisms (invertebrates and vertebrates). Vertebrate cells may be used as host cells.
  • mammalian cell lines that have been adapted for growth in suspension may be used.
  • useful mammalian host cell lines are SV40 transformed monkey kidney CV1 line (COS-7) and human embryonic kidney line (293HEK or 293 cells), etc.
  • Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR - CHO cells and myeloma cell lines such as Y0, NS0 and Sp2/0.
  • a method of preparing an anti-ANG-2 antibody comprises culturing a host cell comprising a nucleic acid that encodes the antibody, as provided hereinbefore, under conditions suitable for expressing the antibody, and optionally recovering the antibody from the host cell (or the culture medium of the host cell).
  • nucleic acids encoding antibodies such as those described hereinbefore, are isolated and inserted into one or more vectors for further cloning and/or expression in host cells.
  • nucleic acids can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes capable of specifically binding to genes encoding the heavy and light chains of the antibodies).
  • anti-ANG-2 antibodies provided herein can be identified, screened, or characterized for physical/chemical properties and/or biological activities by a variety of assays known in the art.
  • an antibody of the invention is tested for antigen-binding activity.
  • the binding of the antibody to human ANG-2 can be determined e.g., by methods known in the art, such as ELISA, Western blotting, or by exemplary methods disclosed herein in the Examples, e.g., as described in Example 3 or 4.
  • the binding profile of the antibody including binding kinetics (e.g., K D value) can be determined using a recombinant ANG-2 protein or a C-terminal FLD fragment thereof in a biological optical interferometry assay.
  • Fortebio affinity assay is employed to determine the binding affinity of the antibody to ANG2 and/or ANG1 derived from human or other species, such as cynomolgus monkeys.
  • the blocking effect of an antibody on ANG2/TIE2 binding or ANG2/integrin binding is assayed.
  • the determination can be performed using e.g., a flow cytometry assay in which the antibody reacts with ANG2 and TIE2 or integrin expressing cell lines , such as 293 cells transfected to express human TIE2 on the cell surface or U87 cells that express a number of integrins.
  • competition assays can be used to identify antibodies that compete with any of the anti-ANG-2 antibodies disclosed herein for binding to ANG-2.
  • such competitive antibodies bind to epitopes (e.g., linear or conformational epitopes) that are identical to or overlapping with the epitope bound by any anti-Ang-2 antibody disclosed herein.
  • the present invention also provides assays for determining the biological activities of anti-ANG-2 antibodies.
  • Biological activities may include, for example, binding to ANG-2 (e.g., binding to human ANG-2), blocking the binding of ANG-2 (e.g., binding of human ANG-2) to Tie2 molecules or integrins, and when in combination with an anti-VEGF agent, inhibiting vascular permeability and/or neovascularization in for example a choroidal neovascularization model, and improving the integrity of blood vessels.
  • Various exemplary assays are described in the Examples of the present invention.
  • any of the above assays can be performed using the immunoconjugates or multispecific antibodies of the present invention, in lieu of or in addition to the anti-ANG-2 antibodies.
  • the present invention provides multispecific (including bispecific) antibody molecules that specifically bind to ANG-2, preferably human ANG-2.
  • the multiple-specific antibody comprises an antibody (or antigen-binding fragment thereof) of the present invention that provides a first binding specificity for ANG-2.
  • the multispecific antibody further comprises a second specificity for VEGF-A.
  • binding specificities are provided by the "binding site” or "antigen-binding site” of an antibody (i.e., the region in the antibody molecule that actually binds to the antigen).
  • the antigen-binding site is composed of a VH/VL pair consisting of an antibody light chain variable domain (VL) and an antibody heavy chain variable domain (VH).
  • VL antibody light chain variable domain
  • VH antibody heavy chain variable domain
  • “multispecific" antibodies have at least two antigen-binding sites, each of which binds to a different epitope on the same antigen or a different epitope on a different antigen.
  • the present invention provides immunoconjugates formed by conjugating an antibody of the invention to a heterologous molecule.
  • the antibody (or antigen-binding fragment thereof) of the invention in the immunoconjugate is conjutated to a therapeutic or diagnostic agent.
  • the antibody of the invention that is conjugated to the heterologous molecule may be a full-length antibody or an antibody fragment.
  • the antibody may be conjugated in the form of a fragment, such as Fab fragment, Fab' fragment, F(ab)' 2 fragment, single-chain scFab antibody and single-chain scFv.
  • Linkers can be used to covalently link different entities of the conjugates. Suitable linkers include chemical linkers or peptide linkers.
  • the linker is a "cleavable linker" that facilitates the polypeptide release upon delivery to a target site.
  • acid-labile linkers, peptidase-sensitive linkers, photolabile linkers, dimethyl linkers, or disulfide-containing linkers can be used.
  • an antibody of the invention can be conjugated to a diagnostic or detectable agent.
  • conjugates can be used as part of a clinical test method (e.g., to determine the efficacy of a particular therapy), or used for monitoring or predicting the onset, development, progression, and/or severity of a disease or a disorder.
  • the antibody can be conjugated to a detectable agent, including, but not limited to, a variety of enzymes, e.g., but not limited to, horseradish peroxidase; prosthetic groups, e.g., but not limited to, streptavidin/biotin and avidin/biotin; fluorescent substances; luminescent substances; radioactive materials; as well as positron-emitting metals and non-radioactive paramagnetic metal ions used in various positron emission tomography imaging procedures.
  • a detectable agent including, but not limited to, a variety of enzymes, e.g., but not limited to, horseradish peroxidase; prosthetic groups, e.g., but not limited to, streptavidin/biotin and avidin/biotin; fluorescent substances; luminescent substances; radioactive materials; as well as positron-emitting metals and non-radioactive paramagnetic metal ions used in various positron emission tomography imaging procedures.
  • enzymes
  • compositions comprising anti-ANG-2 antibodies or immunoconjugates or multispecific antibodies thereof and compositions comprising polynucleotides encoding anti-ANG-2 antibodies or immunoconjugates or multispecific antibodies thereof.
  • compositions can optionally further comprise suitable pharmaceutical auxiliary materials, such as pharmaceutical carriers and pharmaceutical excipients (including buffers) known in the art.
  • Antibodies of the invention can be formulated in any suitable dosage form. In some embodiments, antibodies of the invention are formulated into a formulation suitable for intraocular administration, such as a formlation suitable for intravitreal injection administration. In other embodiments, antibodies of the invention are formulated into a formulation suitable for intravenous injection administration.
  • Pharmaceutical carriers suitable for formulating the formulations of the invention may include, but not limited to, buffers, antioxidants, preservatives and stabilizers.
  • a pharmaceutical formulation of the invention can be formulated, preferably in the form of a lyophilized formulation or an aqueous solution, by mixing an anti-ANG-2 antibody, an immunoconjugate or a multispecific antibody of the invention having a desired purity with one or more optional pharmaceutical excipients ( Remington's Pharmaceutical Sciences, 16th ed., Osol, A. Ed. (1980 )).
  • sustained release formulations can also be prepared.
  • sustained-release formulations include semi-permeable matrices of solid hydrophobic polymers containing antibodies, in the form of shaped articles, such as films or microcapsules.
  • a pharmaceutical composition or formulation of the invention can also be combined with additional active agent(s) which is required for the specific indication being treated, preferably those active agents having complementary activities that do not adversely affect each other.
  • additional active agent(s) which is required for the specific indication being treated, preferably those active agents having complementary activities that do not adversely affect each other.
  • additional active agent(s) preferably those active agents having complementary activities that do not adversely affect each other.
  • additional active agent(s) preferably those active agents having complementary activities that do not adversely affect each other.
  • additional active agent(s) which is required for the specific indication being treated, preferably those active agents having complementary activities that do not adversely affect each other.
  • additional active agent(s) which is required for the specific indication being treated, preferably those active agents having complementary activities that do not adversely affect each other.
  • anti-cancer active agents such as chemotherapeutic agents, PD-1 axis binding antagonists (e.g., anti-PD-1 antibody or anti-PD-L1 antibody or anti-PD-L2 antibody
  • the present invention also provides pharmaceutical combination products.
  • the combination product comprises an antibody, an immunoconjugate, or a multispecific antibody of the invention and a second therapeutic agent, which are formulated in the same pharmaceutical composition or formulation.
  • the combination product comprises an antibody, an immunoconjugate or a multispecific antibody of the invention and a second therapeutic agent, which are separately contained in different pharmaceutical compositions or formulations.
  • the second therapeutic agent can be administered prior to, simultaneously with (e.g., in the same formulation or in a different formulation) or after administration of the antibody of the invention.
  • the second therapeutic agent suitable for use in the present invention may be any therapeutic agent advantageously used in combination with an ANG2 inhibitor, such as a drug that is remedial to a vascular-related disease to be treated.
  • Anti-VEGF agents for angiogenesis include, for example, monoclonal antibodies that specifically bind to VEGF-A, which mechanism of action is to block the binding of VEGF to its receptor by binding to VEGF; VEGFR blocking agents consisting of an extracellular fragment of VEGF receptor and an Fc region of an immunoglobulin, which have an inherently high specificity for VEGF.
  • an effective anti-VEGF agent molecule which is composed of the second immunoglobulin-like domain of VEGFR-1, the third immunoglobulin-like domain of VEGFR-2, and a human immunoglobulin Fc, wherein a peptide linker that increases the structural flexibility is inserted before the Fc, thereby improving the stability and the biological activity of the fusion protein, so that the fusion protein can bind and neutralize VEGF, block angiogenesis, and inhibit tumor progression more effectively, both in vitro and in animals.
  • the second therapeutic agent for use in combination with the antibody of the present invention is an anti-VEGF agent.
  • the anti-VEGF agent is a fusion protein formed from the second immunoglobulin-like domain of VEGFR-1, the third immunoglobulin-like domain of VEGFR-2, and a human immunoglobulin Fc, wherein a peptide linker that increases the structural flexibity is inserted before the Fc, such as the molecule shown in SEQ ID NO: 60, or a molecule having at least 90%, 91%, 92%,93%, 94%, 95%, 96%, 97%, 98%, or 99% identity thereto.
  • anti-VEGF agents such as VEGF-Traps (see, e.g., US7,087,411 ), anti-VEGF antibodies (e.g., bevacizumab), small molecule kinase inhibitors for VEGF receptors (e.g., sunitinib) may also be used in the present invention.
  • VEGF-Traps see, e.g., US7,087,411
  • anti-VEGF antibodies e.g., bevacizumab
  • small kinase inhibitors for VEGF receptors e.g., sunitinib
  • Second therapeutic agents that can be used in the present invention also include, but not limited to, other drugs that inhibit or reduce angiogenesis, anti-inflammatory drugs, immune checkpoint drugs such as anti-PD1 and anti-CD40 drugs, and chemotherapeutic drugs.
  • compositions or pharmaceutical formulations and combination products of the invention are suitable for the prevention or treatment of vascular-related diseases, including diseases or disorders caused by or associated with angiogenesis and/or vascular permeability.
  • compositions, pharmaceutical formulations, and combination products of the invention may be provided as articles of manufacture for the treatment, prevention, and/or diagnosis of the diseases and/or disorders described herein.
  • Such articles of manufacture may comprise a container and a label or a package insert. Suitable containers include bottles, syringes, IV infusion bags, to name a few. Such containers can be made of a variety of materials such as glass or plastic.
  • the articles of manufacture may comprise (a) a first container containing an antibody or the fragment thereof, an immunoconjugate, or a multispecific antibody of the invention, and optionally (b) a second container containing a second therapeutic agent.
  • the articles of manufacture may contain other desirable materials from a commercial and user's view, including buffers, pharmaceutically acceptable diluents such as sterile water for injection, needles, syringes, and syringe pumps.
  • mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • domesticated animals e.g., cows, sheep, cats, dogs, and horses
  • primates e.g., humans and non-human primates such as monkeys
  • rabbits e.g., mice and rats
  • rodents e.g., mice and rats
  • treatment refers to a clinical intervention intended to alter the natural course of a disease in an individual being treated.
  • the desired therapeutic effects include, but are not limited to, preventing disease onset or recurrence, alleviating symptom, minimizing any direct or indirect pathological consequences of disease, preventing metastasis, postponing disease progression, improving or relieving disease status, as well as alleviating or improving prognosis.
  • the present invention provides a method of inhibiting a biological activity mediated by ANG-2, including administering a therapeutic effective amount of an antibody of the invention.
  • the present invention relates to a method of preventing or treating a disease or disorder associated with an ANG2 activity by using an antibody of the invention.
  • the ANG2 activity-associated diseases or disorders includes any diseases or disorders that can be ameliorated, alleviated, prevented, or treated by eliminating, inhibiting, or reducing ANG2 activity.
  • the antibody of the invention is used to treat vascular-related diseases, including diseases or disorders caused by, or associated with, angiogenesis. Examples of such diseases include, but are not limited to, cancers, inflammatory conditions, sepsis, and ocular vascular diseases.
  • an antibody of the invention is used to treat solid tumors.
  • the antibody of the invention can be used alone or in combination with additional antitumor agent(s).
  • an antibody of the invention is used to treat ocular vascular diseases.
  • ocular diseases may be caused by abnormal angiogenesis or vascular obstruction or occlusion.
  • ocular diseases that can be treated with the antibody of the invention, particularly in combination with anti-VEGF agents, include, but are not limited to, ocular neovascularization diseases, and ocular diseases associated with choroidal neovascularization, retinal edema and inflammation.
  • an antibody of the invention when used in combination with an anti-VEGF agent for a vascular-related disease, such as an ocular disease, e.g., choroidal neovascularization (CNV), results in one or more of reduction of vascular leakage, inhibition of neovascularization, and improvement of vascular integrity.
  • a vascular-related disease such as an ocular disease, e.g., choroidal neovascularization (CNV)
  • CNV choroidal neovascularization
  • vascular leakage As revealed by assessment of vascular leakage, retinal morphology, neovascularization and vascular integrity e.g., via fundus fluorescein angiography, or pathological examination based on histochemical staining (e.g., conventional HE staining can be used to examine retinal morphology such as the size of injured and swollen area, and double-fluorescence staining for CD31 and Desmin can be used to evaluate vascular endothelial cell number and pericyte/smooth muscle cell number), preferably, as compared to the anti-VEGF agent alone, the antibody of the present invention in combination with the anti-VEGF agent results in a further improved therapeutic effect, for example, improved vascular leakage inhibition, improved retinal morphology, reduced neovascularization and improved vascular integrity.
  • histochemical staining e.g., conventional HE staining can be used to examine retinal morphology such as the size of injured and swollen area, and double-flu
  • an antibody of the invention or antibody fragment thereof is used to treat infectious diseases.
  • infectious diseases include, but are not limited to, sepsis and pneumonia.
  • the antibodies of the invention can be administered alone or in combination with a second therapeutic agent, such as a second therapeutic agent described hereinbefore.
  • a second therapeutic agent such as a second therapeutic agent described hereinbefore.
  • the combined administration includes concomitant administration (wherein two or more therapeutic agents are contained in the same or different formulations) and separate administration (wherein the antibodies of the invention are administered before, simultaneously with, or after the administration of additional therapeutic agent(s)).
  • the antibodies of the invention can be administered via any suitable administration mode, including parenteral administration, intrapulmonary administration, and intranasal administration as well as intralesional administration if local treatment is required.
  • Parenteral infusions include intramuscular, intravenous, intra-arterial, intraperitoneal or subcutaneous administration. Any suitable route, such as injection, e.g., intravenous or subcutaneous injection, can be adopted, partially depending on whether the administration is a short-time one or a long-term one.
  • the present disclosure encompasses a variety of dosing courses, including, but not limited to, single dosing or multiple dosing at multiple time points, bolus dosing, and pulsed infusion.
  • an antibody of the invention when used alone or in combination with one or more additional therapeutic agents, will be selected depending on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for prophylactic or therapeutic purpose, previous treatment, the patient's clinical history and response to the antibody treatment, as well as the judgment of the attending physician.
  • the antibody is suitably administered to the patient in a single treatment or over a series of treatments.
  • compositions, a multispecific antibody or an immunoconjugate of the invention may be administered in lieu of an antibody or antigen-binding portion of the invention.
  • the composition, multispecific antibody or immunoconjugate of the invention in addition to administering the antibody or antigen-binding portin of the invention, can be further administered.
  • the present invention provides the uses of anti-ANG-2 antibodies, compositions, immunoconjugates, and multispecific antibodies of the invention in the manufacture of a medicament for use in the foregoing methods (e.g., for treatment).
  • the present invention provides methods of regulating, especially reducing, ANG-2-induced vascular permeability by using the anti-ANG2 antibodies or antigen-binding fragments thereof of the invention.
  • the methods can be an in vivo or in vitro method.
  • the said methods are an in vitro method.
  • the methods can be used to screen for molecules that regulate (e.g., decrease, e.g., synergistically or additively decrease) vascular endothelial permeability when in combination with the anti-ANG2 antibodies of the invention.
  • the said candidate molecules can be peptides or small molecule compounds, such as ANG1 analogues, VEGF inhibitors, and the like.
  • the said methods are an in vivo method comprising administering to an individual in need thereof a therapeutically effective amount of an antibody or antigen-binding fragment thereof of the invention.
  • the said methods are used to prevent, alleviate, ameliorate, and/or treat a disease or a disorder associated with abnormal increase of vascular permeability (e.g., acute pneumonia, acute lung injury, and acute respiratory distress syndrome).
  • a disease or a disorder associated with abnormal increase of vascular permeability e.g., acute pneumonia, acute lung injury, and acute respiratory distress syndrome.
  • the abnormal increase of vascular endothelial permeability is associated with VEGF and can be sensitized by ANG-2.
  • the abnormal increase of vascular endothelial permeability is caused by an inflammatory agent such as LPS or a bacterial or viral infection.
  • the said methods are used to alleviate the abnormal increase of vascular endothelial permeability in the lung.
  • the said methods further comprise the step of identifying a subject in need of reducing vascular permeability prior to administration of the anti-ANG2 antibody of the invention.
  • the expression of an inflammatory marker molecule on a vascular endothelial cell is reduced by the administration of an anti-ANG2 antibody of the invention.
  • the said inflammatory marker molecule is a molecular marker of acute inflammatory response.
  • increased expression of the inflammatory marker on vascular endothelial cells is induced in response to an inflammatory cytokine such as TNF- ⁇ or a inflammatory agent such as the bacterial endotoxin LPS.
  • the inflammatory marker molecule is ICAM-1 and/or VCAM-1.
  • the said methods further comprise the step of detecting the level of an inflammatory marker molecule on endothelial cell prior to the administration of the anti-ANG2 antibody of the invention.
  • the subject to be administered with the antibody of the invention has an elevated level of an inflammatory marker molecule on endothelial cell, such as ICAM-1 and/or VCAM-1.
  • the subject to be administered with the antibody of the invention is an individual at high risk for ARDS or acute lung injury and having an elevated level of an inflammatory marker molecule on endothelial cell, such as ICAM-1 and/or VCAM-1.
  • the present invention relates to a method of preventing or treating vascular leakage, comprising administering to a subject in need thereof a therapeutically effective amount of an antibody of the invention or an antigen-binding fragment thereof.
  • an ANG2 activity is blocked, inhibited, or reduced by administration of the anti-ANG2 antibody of the invention, thereby ameliorating, alleviating, preventing, or treating vascular leakage or one or more conditions associated therewith.
  • the said method further comprises the step of identifying a subject in need of blocking vascular leakage prior to the administration of the anti-ANG2 antibody of the invention.
  • the present invention relates to a method of preventing or treating a vascular inflammatory condition, comprising administering to a subject in need thereof a therapeutically effective amount of an antibody of the invention or an antigen-binding fragment thereof.
  • an ANG2 activity is blocked, inhibited, or reduced by administration of the anti-ANG2 antibody of the invention , thereby ameliorating, alleviating, preventing, or treating the vascular inflammatory condition.
  • the said method further comprises the step of identifying a subject in need of alleviating a vasulcar inflammatory condition prior to the administration of the anti-ANG2 antibody of the invention.
  • the invention also relates to a method of preventing or treating a disease or a disorder accompanied with vascular leakage and/or vascular inflammatory condition, the said method comprising administering to a subject in need thereof a therapeutically effective amount of an antibody of the invention or an antigen-binding fragment thereof.
  • the disease or disorder is a disease or disorder accompanied with vascular leakage.
  • the disease or disorder is a disease or disorder accompanied with vascular inflammation. Examples of such diseases or conditions include, but are not limited to, pulmonary inflammatory conditions such as pneumonia, acute lung injury (ALI), and acute respiratory distress syndrome (ARDS) and sepsis.
  • Pulmonary inflammation or pneumonia is defined as a disease characterized by inflammation of the lung. Most commonly, the disease is caused by a lung infection.
  • the pneumonia is acute pneumonia, especially acute pneumonia at risk of developing ARDS or acute lung injury (ALI).
  • the administration of an anti-ANG2 antibody blocks, prevents, alleviates or reduces the inflammatory process in the lung. The improvement of the inflammatory process in the lung can be observed by lung biopsy or lung CT images.
  • Acute lung injury is an acute hypoxic respiratory insufficiency caused by diffuse pulmonary interstitial and alveolar edema due to the damage of alveolar epithelial cells and capillary endothelial cells induced by various direct and indirect injury factors.
  • the pathophysiological features of ALI include reduced lung volume, reduced lung compliance, and ventilation/blood flow imbalance. Clinically, it is characterized by progressive hypoxemia.
  • Acute respiratory distress syndrome (ARDS) may occur when the injury reaches a certain degree.
  • Acute respiratory distress syndrome is also called ARDS.
  • the disease is a type of acute lung injury, characterized by extensive lung inflammation. Patients with ARDS usually manifest with bilateral lung infiltration and severe hypoxemia.
  • the diagnostic criteria of ARDS includes time of onset; chest imaging (such as X-ray or CT scanning of the chest); pulmonary edema; and the degree of hypoxia.
  • Hypoxemia for the diagnosis of ARDS can be defined, for example, by the oxygenation index of the patient's arterial blood (PaO 2 /FiO 2 ; i.e., the ratio of the partial pressure of arterial oxygen to the fraction of inspired oxygen).
  • ARDS In the 2012 Berlin definition of ARDS, based on the degree of hypoxemia determined by PaO2/FiO2, ARDS is classified into three categories: mild ARDS (PaO 2 /FiO 2 200-300mm Hg), moderate ARDS (PaO 2 /FiO 2 100-200 mm Hg), and severe ARDS (PaO 2 /FiO 2 ⁇ 100 mm Hg) (see, e.g., The ARDS Definition Task Force, JAMA 307 (23): 2526-2533, 2012 ).) As will be understood, in the present disclosure, ARDS covers any appropriate ARDS clinical definitions known in the art, including the 2012 Berlin definition. In the present disclosure, individuals at risk for ARDS refer to those who are susceptible to ARDS. Trauma, major bleeding, severe pneumonia, influenza or SARS/COVID-19 infection, and/or sepsis are the risk factors for ARDS. Individuals with these risk factors may be individuals at risk for ARDS.
  • a pulmonary inflammatory condition such as ARDS, ALI or pneumonia
  • the infection can be a bacterial infection, a viral infection, a fungal infection, a parasitic infection, or an infection by other types of microorganisms.
  • the bacterial infection is caused by Enterobacteriaceae species, Streptococcus pneumoniae, Staphylococcus aureus, Bacillus anthracis, Haemophilus influenzae, Klebsiella pneumoniae, Escherichia coli, Pseudomonas aeruginosa, Bordetella pertussis, Chlamydia pneumoniae, Legionella spp. or Mycoplasma pneumoniae.
  • the viral infection is caused by influenza virus, parainfluenza virus, respiratory syncytial virus, human parainfluenza virus, adenovirus, metapneumovirus, severe acute respiratory syndrome (SARS) virus, herpes simplex virus (HSV), varicella zoster virus (VZV), measles virus, rubella virus, cytomegalovirus (CMV), variola virus, dengue virus, rhinovirus, bocavirus, Middle East respiratory syndrome virus, or coronavirus.
  • SARS severe acute respiratory syndrome
  • HSV herpes simplex virus
  • VZV varicella zoster virus
  • measles virus rubella virus
  • CMV cytomegalovirus
  • variola virus dengue virus
  • rhinovirus bocavirus
  • Middle East respiratory syndrome virus or coronavirus.
  • coronavirus One of the major host responses to infection is inflammatory response, which leads to pulmonary microvascular leakage and lung injury, sometimes may further lead to respiratory failure.
  • an anti-ANG2 antibody or pharmaceutical composition thereof of the present invention is administered prior to appearance of a clinical symptom of an infectious disease.
  • the anti-ANG2 antibody of the present invention, or pharmaceutical composition thereof can be administered after an infectious agent (e.g., bacteria or viruses) is detected present in a sample from a patient or after a patient is exposed to an infectious agent (e.g., bacteria or viruses, including in close contact with an asymptomatic and/or symptomatic infected individual).
  • an infectious agent e.g., bacteria or viruses
  • an infectious agent e.g., bacteria or viruses
  • the invention relates to a method of preventing or treating acute respiratory distress syndrome by using an antibody of the invention; or a method of ameliorating one or more parameters of acute respiratory distress syndrome by using an antibody of the invention, wherein the method comprises administration to a subject in need thereof a therapeutically effective amount of an antibody or antigen-binding fragment thereof of the invention.
  • the said parameter is selected from the group consisting of: infiltration intensity of inflammatory cells in lung tissues (including, but not limited to, leukocyte count and differential inflammatory cell count in lung lavage fluid, such as neutrophil count, lymphocyte count and monocyte count), intensity of inflammatory lesions in lung tissues (including but not limited to, lesion intensity grades), arterial oxygen saturation (including, but not limited to, PO 2 , PCO 2 , pH, sO 2 % value), lung images (X-ray or CT images), degree of pulmonary edema, lung weight index, and/or survival rate, or any combination thereof.
  • the said parameter comprises the infiltration intensity of inflammatory cells in the lung.
  • the said parameter comprises arterial oxygen saturation.
  • the said method further comprises, prior to the administration of the anti-ANG2 antibody of the invention, identifying an individual eligible for prevention or treatment of acute respiratory distress syndrome or for ameliorating one or more of the aforementioned parameters of ARDS.
  • the subject has mild, moderate, or severe hypoxemia.
  • the subject is at risk of developing hypoxemia, and the anti-ANG2 antibody of the invention is administered prophylactically.
  • the subject has suffered from lung inflammation and presented with at least a slight, mild, moderate or severe inflammatory lesions in lung tissues.
  • the subject is at risk of developing an inflammatory lesion in lung tissues, and the anti-ANG2 antibody of the invention is administered prophylactically.
  • the intensity of inflammatory lesions in the lung tissues can be evaluated, for example, by examining the infiltration intensity of inflammatory cells in alveoli/interstitium/bronchi/pleura, the degree of pulmonary edema/fibrin-like material exudation, the degree of thrombus, the degree of degeneration/necrosis in blood vessels/bronchi/alveoli/interstitium, the degree of abscess, the degree of alveolar wall thickening, and the degree of alveolar/pleural hemorrhage.
  • Oxygen saturation can be evaluated in a manner known in the art.
  • PO 2 , PCO 2 , pH, and sO 2 % values can be measured using collected arterial blood with a blood gas analyzer to detect (e.g., before and after administration of the antibody of the invention) the hypoxemic condition of the subject.
  • the methods of the invention include treating one or more symptoms of ARDS, such as mild, moderate, or severe hypoxemia.
  • a subject with hypoxemia may exhibit alterations in one or more parameters, such as partial pressure of oxygen, oxygenation index (PaO 2 /FiO 2 , i.e., the ratio of arterial partial pressure of oxygen to the fraction of inspired oxygen), CT images of the lung (e.g., localized lung inflammation, increased markings in both lungs, decreased transmittance of both lungs), and levels of one or more inflammatory markers, relative to a healthy subject.
  • the present invention also relates to a use of an anti-ANG2 antibody of the invention as a medicament or in the manufacture of a medicament, wherein the medicament may be used in any one or more of the above methods of the invention.
  • the anti-ANG2 antibody is used to modulate, especially reduce, the ANG-2-induced vascular endothelial permeability.
  • the anti-ANG2 antibody is used to prevent or treat vascular leakage and/or vascular inflammatory condition associated with ANG2 activity.
  • the anti-ANG2 antibody is used to prevent or treat a disease or disorder accompanied with vascular leakage and/or vascular inflammatory condition.
  • the said disease or condition is selected from the group consisting of: pulmonary inflammatory condition (particularly acute inflammatory condition), sepsis, acute lung injury, and acute respiratory distress syndrome.
  • the anti-ANG2 antibodies of the invention may bind to ANG2 on endothelial cells of a subject and block the binding of ANG2 to its receptor (e.g., TIE2 or an integrin).
  • the anti-ANG2 antibodies of the invention block the interaction of ANG2 with TIE2.
  • the anti-ANG2 antibodies of the invention block the interaction of ANG2 with an integrin.
  • the administration of the anti-ANG2 antibodies inhibits the expression of an inflammatory marker on an endothelial cell.
  • the administration of the anti-ANG2 antibodies inhibits an increase in vascular permeability (caused by e.g., an inflammatory agent such as a cytokine or LPS).
  • the antibodies of the invention can be administered alone or in combination with a second therapeutic agent.
  • the antibodies of the invention are administered as the sole active agent.
  • the antibodies of the invention are administered in combination with additional therapeutic agent(s), wherein the said combined administration comprises concomitant administration (in which the two or more therapeutic agents may be contained in the same or different formulations) and separate administration, wherein the antibodies of the invention can be administered before, simultaneously with, or after the administration of the additional therapeutic agent (s).
  • the second therapeutic agent may be, for example, an anti-inflammatory agent, such as a glucocorticoid (e.g., dexamethasone); an anti-infective agent (e.g., antibiotics, antivirals, antifungals, antiparasitics); a bronchodilator (such as ⁇ 2 agonists); a vasopressor (e.g., isoproterenol, phenylephrine); or a sedative (e.g., propofol, escitalopram, morphine).
  • an anti-inflammatory agent such as a glucocorticoid (e.g., dexamethasone); an anti-infective agent (e.g., antibiotics, antivirals, antifungals, antiparasitics); a bronchodilator (such as ⁇ 2 agonists); a vasopressor (e.g., isoproterenol, phenylephrine
  • the subject was a mammal. In preferable embodiments, the subject was human. In some embodiments, the subject is identified as an individual in need of treatment by detection of one or more of the following: inflammatory marker level in vascular endothelium, arterial blood gas level, partial pressure of oxygen (PaO 2 ) level, partial pressure of carbon dioxide (PaCO 2 ) level, the intensity of pulmonary inflammatory infiltration and chest imaging.
  • inflammatory marker level in vascular endothelium arterial blood gas level
  • PaO 2 partial pressure of oxygen
  • PaCO 2 partial pressure of carbon dioxide
  • composition of the invention may be administered in lieu of the antibody or antigen-binding portion of the invention.
  • composition of the invention in addition to administering the antibody or antigen-binding portion of the invention, can be further administered.
  • the antibodies of the invention are administered in the form of pharmaceutical compositions.
  • the pharmaceutical compositions of the present invention can be formulated using suitable carriers, excipients, and other substances that provide suitable transfer, delivery, tolerance, and the like.
  • suitable formulations can be found in a pharmacopoeia known to all medicinal chemists: Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, PA .
  • formulations include, for example, powders, pastes, ointments, gels, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTIN TM ), DNA conjugates, anhydrous absorbent pastes, oil-in-water and water-in-oil emulsions, carbowaxes (polyethylene glycols of various molecular weights) emulsions, semi-solid gels, and semi-solid mixtures containing carbowaxes.
  • vesicles such as LIPOFECTIN TM
  • DNA conjugates such as LIPOFECTIN TM
  • anhydrous absorbent pastes oil-in-water and water-in-oil emulsions
  • carbowaxes polyethylene glycols of various molecular weights
  • the dosage of antibody administered to a patient according to the methods of the invention may vary depending on the patient's age and body size, symptoms, conditions, route of administration, and the like. Usually, the dosage is calculated based on body weight or body surface area. The frequency and duration of treatment can be adjusted depending on the severity of the disease.
  • the effective dosage and dosing regimen for administering a pharmaceutical composition comprising an anti-ANG2 antibody can be determined empirically. For example, disease progression can be monitored through periodic assessment, and the dosage adjusted accordingly. In addition, inter-species scaling of dosage can be performed using methods well known in the art.
  • a variety of delivery systems are known in the art and can be used to administer pharmaceutical compositions comprising the anti-ANG2 antibodies of the present invention, such as syringes, pre-filled syringes, auto-injectors, microinfusors, glass vials, encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing mutant viruses and receptor-mediated endocytosis.
  • Adminstration methods include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • composition can be administered by any convenient route of administration, such as by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, and the like), and can be co-administered with other biologically active agents.
  • routes of administration such as by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, and the like), and can be co-administered with other biologically active agents.
  • the pharmaceutical compositions of the present invention can be delivered subcutaneously or intravenously using standard needles and syringes.
  • the syringe is a pre-filled syringe.
  • Such a pre-filled syringe can be a single-dose syringe.
  • the pharmaceutical composition of the present invention is delivered subcutaneously or intravenously with an auto-injector, wherein the auto-injector may contain a pre-filled cartridge.
  • pen delivery devices can be readily used to deliver the pharmaceutical compositions of the present invention.
  • Such a pen delivery device can be reusable or disposable.
  • a reusable pen delivery device typically utilizes a replaceable cartridge containing a pharmaceutical composition.
  • the empty cartridge can be easily discarded and replaced with a new cartridge containing the pharmaceutical composition.
  • the pen delivery device can then be used again.
  • the disposable pen delivery device is prefilled with a pharmaceutical composition contained in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded.
  • a number of reusable syringes, pens, and autoinjector delivery devices can be used to deliver the pharmaceutical compositions of the present invention subcutaneously. Examples include, but are not limited to, AUTOPEN TM (Owen Mumford, Inc., Woodstock, UK), DISETRONIC TM Pen (Disetronic Medical Systems, Bergdorf, Switzerland), HUMALOG MIX 75/25 TM Pen, HUMALOG TM Pen, HUMALIN 70/30 TM Pen (Eli Lilly and Co., Indianapolis, IN), NOVOPEN TM I, II and III (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIOR TM (Novo Nordisk, Copenhagen, Denmark), BD TM Pen (Becton Dickinson, Franklin Lakes, NJ), OPTIPEN TM and OPTIPEN PRO TM .
  • SOLOSTAR TM pen Sanofi-Aventis
  • FLEXPEN TM Novo Nordisk
  • KWIKPEN TM Eli Lilly
  • SURECLICK TM auto-injector Amgen, Thousand Oaks, CA
  • PENLET TM Heaselmeier, Stuttgart, Germany
  • EPIPEN Dey, L.P.
  • HUMIRA TM pen Abbott Labs, Abbott Park IL
  • the pharmaceutical composition can be delivered in a controlled release system.
  • a pump can be used.
  • polymeric materials can be used.
  • a controlled release system can be placed proximate to the target of the composition, so that only a portion of the systemic dose is required.
  • the injectable formulation may include dosage forms for intravenous, subcutaneous, intradermal and intramuscular injection, drip infusion, and the like.
  • injectable formulations can be prepared using the methods known in the art.
  • an injectable formulation can be prepared, for example, by dissolving, suspending or emulsifying the antibody or salt thereof described hereinbefore in a sterile aqueous medium or an oily medium conventionally used for injection.
  • aqueous medium for injection there are, for example, physiological saline, isotonic solution containing glucose and other auxiliary agents or the like, which can be used in combination with suitable solubilizers such as alcohols (for example, ethanol), polyols (for example, propylene glycol, polyethylene glycol), nonionic surfactants [for example, polysorbate 80, HCO-50 (polyoxyethylene (50mol) adduct of hydrogenated castor oil)] or the like.
  • suitable solubilizers such as alcohols (for example, ethanol), polyols (for example, propylene glycol, polyethylene glycol), nonionic surfactants [for example, polysorbate 80, HCO-50 (polyoxyethylene (50mol) adduct of hydrogenated castor oil)] or the like.
  • suitable solubilizers such as alcohols (for example, ethanol), polyols (for example, propylene glycol, polyethylene glycol), nonionic surfactants [for example, polysorbate
  • the aforementioned pharmaceutical compositions for oral or parenteral use can be formulated into dosage forms in a unit dose suited to fit a dose of the active ingredient.
  • dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, and the like.
  • the antibodies of the invention can be administered via any suitable administration mode, including parenteral administration, intrapulmonary administration, and intranasal administration as well as intralesional administration, if local treatment is required.
  • Parenteral infusions include intramuscular, intravenous, intra-arterial, intraperitoneal or subcutaneous administration. Any suitable route, such as injection, e.g., intravenous or subcutaneous injection, can be adopted, partially depending on whether the administration is a short-time one or a long-term one.
  • the present disclosure encompasses a variety of dosing courses, including, but not limited to, single dosing or multiple dosing at multiple time points, bolus dosing, and pulsed infusions.
  • an antibody of the invention when used alone or in combination with one or more other therapeutic agents, will be selected depending on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for prophylactic or therapeutic purpose, previous treatment, the patient's clinical history and response to the antibody treatment, as well as the judgment of the attending physician.
  • the antibody is suitably administered to a patient in a single treatment or over a series of treatments.
  • the anti-ANG2 antibodies of the invention are administered subcutaneously or intraperitoneally. In yet some preferable embodiments, the anti-ANG2 antibodies of the invention are administered in single or multiple dosages. Preferably, at least one dosage of the anti-ANG2 antibody of the present invention is administered before or immediately after the subject is exposed to an inflammatory agent, such as a bacterium, virus, bacterial toxin, and more preferably before the subject manifests pulmonary infiltration of inflammatory cells. In some embodiments, the antibodies are formulated as a liquid composition.
  • the antibodies are formulated for inhalation, insufflation, nebulization, or injection administration, or as pharmaceutical compositions for oral, rectal, topical, or intraperitoneal administration.
  • the injection is intravenous injection.
  • the anti-ANG2 antibody of the present invention is contained in a pharmaceutical composition, and optionally the pharmaceutical composition is contained in a container selected from the group consisting of a glass vial, a syringe, a prefilled syringe, an autoinjector, and a microinfusion set.
  • the present invention relates to a pharmaceutical combination or pharmaceutical composition for use in the above method or use comprising an anti-ANG2 antibody of the present invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical combination or pharmaceutical composition further comprises a second therapeutic agent.
  • the present invention relates to a kit for use in the method or use described above, which comprises a container comprising a pharmaceutical composition of the present invention and the instructions for use.
  • the kit further comprises a container containing a second therapeutic agent.
  • the invention relates to a method and kit for detecting ANG-2 in a sample, wherein the method comprises: (a) contacting the sample with an antibody of the invention, or an antigen-binding fragment or immunoconjugate thereof, and (b) detecting the formation of a complex between the said antibody or antigen-binding fragment or immunoconjugate thereof and an ANG-2 protein.
  • the sample is derived from a cancer patient or a patient with an ocular disease.
  • the said sample can be a tissue biopsy specimen, a tissue section, a body fluid such as blood, plasma or serum.
  • the said detection can be conducted in vitro or in vivo.
  • detection when used herein, includes quantitative or qualitative detection, and exemplary detection methods may involve immunohistochemistry, immunocytochemistry, flow cytometry (e.g., FACS), antibody molecule-conjugated magnetic beads, ELISA assays and PCR-techniques (e.g., RT-PCR).
  • exemplary detection methods may involve immunohistochemistry, immunocytochemistry, flow cytometry (e.g., FACS), antibody molecule-conjugated magnetic beads, ELISA assays and PCR-techniques (e.g., RT-PCR).
  • the ANG-2 to be detected is human ANG-2 or cynomolgus monkey ANG-2.
  • an anti-ANG-2 antibody is used to select a subject suitable for treatment with an anti-ANG-2 antibody, e.g., when ANG-2 is a biomarker for selecting the subject.
  • antibodies of the invention are used to diagnose a vascular-related disease such as a tumor or an infectious disease such as sepsis, for example, to evaluate (e.g., to monitor) the treatment or progression, the diagnosis and/or staging of a disease described herein in a subject.
  • labeled anti-ANG-2 antibodies include, but are not limited to, labels or moieties that are detected directly (such as fluorescent labels, chromophore labels, electron dense labels, chemiluminescent labels, and radiolabels), and moieties such as enzymes or ligands that are detected indirectly, for example, by enzymatic reactions or molecular interactions.
  • Exemplary labels include, but are not limited to, radioisotopes 32P, 14C, 125I, 3H, and 131I, fluorophores such as rare earth chelates or luciferin and derivatives thereof, rhodamine and derivatives thereof, dansyl, umbelliferone, luciferases, e.g., firefly luciferase and bacterial luciferase ( U.S. Patent. No.
  • luciferin 2,3-dihydrophthalazine-1,4-dione, horseradish peroxidase (HR), alkaline phosphatase, ⁇ -galactosidase, glucoamylase, lytic enzyme, sugar oxidases, e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase, heterocyclic oxidases such as uricase and xanthine oxidase, and enzymes that oxidize dye precursors using hydrogen peroxide such as HR, lactoperoxidase, or microperoxidase, biotin/avidin, spin labels, phage labels, stable free radicals, and the like.
  • HR horseradish peroxidase
  • alkaline phosphatase alkaline phosphatase
  • ⁇ -galactosidase glucoamylase
  • lytic enzyme sugar oxidases, e.g.
  • mice were immunized with human angiopoietin 2 (Ang2) protein, and then the spleen cells of the mice were fused with myeloma cells to obtain hybridoma cells that express positive antibodies.
  • Ang2 angiopoietin 2
  • Basal medium FBS(Hyclone,SH30084.03) 10% GlutaMAX TM Supplement(Gibco, 35050-079) 1 ⁇ RPMI-1640(Hyclone, SH30809.01) 90%
  • Mouse myeloma SP2/0 cells (ATCC, CRL-1581) were resuspended in 20 ml of basal medium and counted. SP2/0 and spleen cells were mixed in a ratio of 1: 2 to 1: 1 and centrifuged at 1000 rpm for 6 min. After removal of the supernatant, the mixed cells were resuspended in 10 ml of cytofusion buffer (BTXpress, 47-0001). Another 15 ml of cytofusion buffer was added, and after centrifugation at 1000 rpm for 5 min, the supernatant was discarded.
  • cytofusion buffer BTXpress, 47-0001
  • Another 15 ml of cytofusion buffer was added, and after centrifugation at 1000 rpm for 5 min, the supernatant was discarded.
  • the cells were resuspended with an appropriate amount of cytofusion buffer and the mixed cell density was adjusted to 1 ⁇ 10 7 cells/ml.
  • the parameters of the electrofusion instrument were set as follows. Electrofusion was performed by adding 2 ml of the cell suspension to each electrofusion dish. Condition Mouse (SP2/0-ECF-F) Alignment: 60v, 30 sec Membrane breaking: 1500V, 30 ⁇ s, 3X Post-fusion pulse: 60V, 3 sec
  • the cells were allowed to stand for 5 min at room temperature in the electrofusion dish. The cells were transferred into centrifuge tubes and diluted to 1-2 ⁇ 10 4 cells/ml with selection medium (see table below). A 100 ⁇ l of cell suspension was added to each well of a 96-well plate. The selection medium was replaced on Day 7 post-fusion. The screening was performed after 10 days of culture (or longer, depending on cell growth status). Hybridoma cells expressing specific anti-hAng2 antibodies were selected by Elisa assay.
  • the antibodies expressed by the selected hybridoma cells were sequenced by PCR to determine the variable region genes of the antibodies, and the chimeric antibodies were screened, humanized, and affinity matured to obtain the antibody sequences of the invention.
  • amino acid sequences of the CDR regions, light chain variable regions, heavy chain variable regions, light chain and heavy chains of six exemplary antibodies of the invention (chimeric antibodies 81H10, 86E9, 107H7; humanized antibody: hz81H10.4; affinity matured antibodies Amhz81H10.4.30, Amhz81H10.4.49), and the corresponding nucleotide sequences are shown in Table D below.
  • Table D The SEQ ID NOs of the revealant sequences of each of the antibodies involved in the present Invention
  • HC is formed by fusing the VH sequence to the IgG1 portion of SEQ ID NO: 52
  • LC is formed by fusing the VL sequence to the C ⁇ chain of SEQ ID NO: 53.
  • the antibody-expressing CHO-S cell lines were generated using the GS Xceed TM Gene Expression System kit (Lonza) according to the manufacturer's instructions.
  • the DNA sequences for the heavy and light chains of an antibody molecule were first inserted into the same pCHO1.0 plasmid, where the heavy chain was positioned upstream of the light chain. Then the constructed pCHO1.0 plasmid was transferred into the CHO cell line by chemical transfection and electrotransfection method. At 48 hours after transfection, the ForteBio method was used to detect the antibody production so as to determine the transfection efficiency. After two rounds of screening under selection pressure with 10 ⁇ g/ml and 50 ⁇ g/ml puromycin, the cell pool with high antibody expression was obtained. After expanding the cell pool to express the antibody in large amounts, the cell supernatant was collected and purified with Protein A to an antibody purity > 95%.
  • plasmids such as pcDNA3.1 can be used.
  • the cDNAs encoding the heavy and light chains of the antibody were first cloned into the pcDNA3.1 vector.
  • the vectors carrying the heavy chain and light chain of the antibody molecule were transferred into HEK293 cells using a chemical transfection method.
  • the chemical tranfection agent used was PEI (purchased from Polysciences) , and the cultured 293HEK (Invitrogen) cells were transiently transfected according to the procedure provided by the manufacturer. After transfection, the medium was discarded and the cells were diluted to 4 ⁇ 10 6 /ml with fresh EXPI293 medium (Gibco).
  • the equilibrium dissociation constants (K D ) of the above exemplary antibodies of the present invention binding to human Ang2 (h Ang2), cynomolgus monkey Ang2 (cAng2), murine Ang2 (mAng2), and human Ang1 (hAng1) were determined using the biological optical interferometry (ForteBio) assay.
  • the ForteBio affinity assay was performed according to the existing method ( Estep, P et al., High throughput solution-based measurement of antibody-antigen affinity and epitope binning. MAbs, 2013.5 (2): p. 270-8 ). Briefly, the sensor was equilibrated in the assay buffer off-line for 30 min, followed by on-line detection for 60s to establish the baseline. Then the purified antibodies obtained as described above were loaded on-line onto an AHQ sensor (ForteBio) for ForteBio affinity assay.
  • the sensors with loaded antibodies were exposed to 100 nM of hAng2, cAng2, mAng2, and hAng1 antigens for 5 min, and then were transferred into the assay buffer for dissociation for 5 min to determine the dissociation rate.
  • Kinetic analysis was performed using a 1: 1 binding model.
  • Table 1A Affinities of Anti-Ang2 Antibodies Antibody Human Ang2 Cynomolgus monkey Ang2 C-terminal domain (Lys274-Phe495) Murine Ang2 C-terminal domain (Lys275-Phe496) Human Ang1 81H10 6.92E-09 4.43E-08 N.B N.B 86E9 7.63E-09 4.62E-08 N.B N.B 107H7 7.75E-09 2.52E-08 N.B N.B hz81H10.4 1.25E-09 8.83E-08 N.B N.B Amhz81H10.4.30 2.32E-08 N.D N.B N.B Amhz81H10.4.49 6.468E-10 N.D N.B N.B Nesvacumab 1.42E-09 1.04E-09 1.02E-09 N.B *N.B. indicates no binding; N.D. indicates not determined.
  • Hz81H10.4 was selected, and the binding abilities of HZ81H10.4 to ang2 proteins of different species, including human ANG2, rhesus ANG2, rat ANG2, mouse ANG2 and rabbit ANG2, were further investigated using Biacore instrument T200.
  • Channel 2 of Protein G chip was selected to capture the antibody, and flow rate was 10 ⁇ L/min in the assay.
  • the antibody was diluted to 1 ⁇ g/mL and was captured onto channel 2 for 30 s, and channel 1 was the blank control channel.
  • the gradient-diluted Human ANG2, Rhesus ANG2, Rabbit ANG2, Rat ANG2 and Mouse ANG2 were sequentially injected into channels 1 and 2 of the chip, from low to high concentrations (0-32 nM), at a flow rate of 30 ⁇ L/min, with one concentration per cycle, and with 180s association time and 1200s dissociation time at 25 °C.
  • ELISA Assay for binding of anti-hAng2 antibody of the present invention to human Ang2 protein The binding of the respective parental murine hybridoma antibodies of the above exemplary chimeric antibodies 81H10, 86E9, and 107H7 to human hAng2 was measured in an ELISA-based assay.
  • Human ang2 protein (Sino Biological, Cat No.: 10691-H08H) was diluted to 0.25 ⁇ g/ml with PBS, and was coated onto a microtiter plate at 100 ⁇ l/well at 4°C overnight. After three washes with PBST, the plate was blocked with 5% BSA. Gradient diluted antibodies (maximum concentration 20 nM, 1:2 serial dilution) were added to the microtiter plate and incubated for 2 h. After three washes with PBST, a 1: 10000 dilution of anti-mouse IgG, HRP-conjugated antibody (Cell Signaling, Cat. No.
  • the binding of 81H10 and hz81H10.4 of the present invention to human biotin Ang2 protein was measured in an SA-biotin Ang2-based ELISA assay.
  • SA protein (Thermo Fisher) was diluted to 1 ⁇ g/ml with PBS, and was coated onto a microtiter plate at 100 ⁇ l/well at 4°C overnight. After three washes with PBST, the plate was blocked with 5% BSA for 1 h. Biotin Ang2 (R&D, Cat. No. BT623B/CF) was incubated at a concentration of 0.5 ⁇ g/ml for 1 h. Gradient diluted antibodies (maximum concentration 20 nM, 1: 3 serial dilution) were added to the microtiter plate and incubated for 2 h, and the plate was washed three times with PBST.
  • Anti-human IgG, HRP-conjugated antibody (Bethyl, Cat. No. A80-104P) diluted at 1:10000 was added and incubated for 60 min. Then the plate was washed for 6 times with PBST. Chromogenic reaction was performed with 100 ul TMB and terminated with 100 ul 2N sulfuric acid. The reading of OD 450 - OD 620 was taken by microplate reader. The concentration-dependent curve was fitted with GraphPad based on the OD 450 - OD 620 .
  • Nesvacumab and faricimab to block the binding of human Ang2 to hTie2 were determined by ELISA.
  • Faricimab is a bispecific antibody drug undergoing a clinical trial and developed by Roche, which targets both angiopoietin-2 (Ang-2) and VEGF-A.
  • the hTie2 protein (Sino Biological, Cat. No.: 10700-H03H) was resuspended and dissolved in PBS to a concentration of 2 ⁇ g/ml, and then coated onto a microtiter plate overnight. Then the plate was blocked using 5% BSA for 1 h.
  • the biotinylated antigen, recombinant biotinylated hAngiopoietin-2 protein (R&D, Cat. No.: BT623B/CF), was diluted to 600 ng/ml, and was added into the microtiter plate at 50 ⁇ l/well.
  • the antibodies (81H10, 86E9, 107H7, hz81H10.4, Amhz81H10.4.30 and Amhz81H10.4.49) prepared as described above were gradiently diluted with a total of 8 or 12 dilution points starting from the highest concentration, and then added to the microtiter plate at 50 ⁇ l/well, and incubated in PBS on ice for 30 min. Then, 50 ul of antigen solution was added to the microtiter plate, in which the final antigen concentration was 300 ng/ml. The plate containing the antigen-antibody mixture was incubated for 90 min.
  • the absorbance values of OD 450 and OD 620 were read using a microplate reader.
  • Human Tie2-overexpressing expi-293 cells (293-tie2 cells) were generated by transfection with the pCHO1.0 vector (Invitrogen) carrying the human Tie2 gene cloned into the MCS.
  • the antigen hAng2-Fc protein (Sino Biological, Cat No.: 10691-H02H) was diluted to 4 ⁇ g/ml, and added to the plate wells at 50 ⁇ l/well.
  • Antibodies (hz81H10.4 and control antibody Nesvacumab) prepared as described above were diluted at 2-fold with a total of 12 dilution points starting from the highest concentration of 800 nM, and added to the plate wells at 50 ⁇ l/well, and incubated in PBS on ice for 30 min, where the final concentration of the antigen was 2 ⁇ g/ml and the highest concentration of antibody was 400 nM.
  • the 293-tie2 cells were adjusted to 2 ⁇ 10 5 cells/well and added to the plate wells at 100 ⁇ l/well. The cells were centrifuged at 300 g for 5 min, and after the supernatant was discarded, the cells were resuspended in the antigen-antibody mixture.
  • the cells were incubated on ice for 30 min, and added with PBS at 100 ⁇ l/well, centrifuged at 300 g for 5 min, and washed with PBS once.
  • a 100 ⁇ l of goat anti-human IgG-PE (Southern Biotech, Cat. No. 2040-09) diluted at 1: 200 was added to each well and placed in an ice bath for 20 min.
  • PBS was added at 100 ⁇ l/well, and the cells were centrifuged at 300 g for 5 min, and washed once with PBS.
  • the cells were resuspended in 100 ⁇ l PBS and the fluorescence signals of the cells were detected by a flow cytometer (BD Biosciences). Concentration-dependent curves were fitted with GraphPad according to their MFI. As shown in Figure 3 , hz81H10.4 had better IC50 values compared to the control antibody Nesvacumab.
  • the expi293 cells overexpressing tie2, i.e., 293-tie2 cells, were incubated with the antibody and recombinant hAng2-Fc protein.
  • the inhibitory effect of the antibody on the phosphorylation of Tie2 induced by hAng2-fc was reflected by the amount of phosphorylated Tie2 detected in the system.
  • Tie2-overexpressing 293-tie2 cells were pipetted and diluted to 2 ⁇ 10 6 cell/ml, and were added at 100 ⁇ l/well into a 96-well plate, then centrifuged at 400 g for 5 min, and the supernatant was removed.
  • Expi293 medium Gibco
  • the test antibody was added to the plate wells (the highest final concentration at 60 ⁇ g/ml, and 1: 2 serial dilution). Then hAng2-Fc was added at a final concentration of 2.5 ⁇ g/ml.
  • the cells were resuspended with 100 ⁇ l/well of the experimental medium and incubated at 37°C for 15 min.
  • the pTie2 concentration was determined according to the protocol of the phospho-tie2 ELISA kit (R&D, Cat. No. DYC2720E). Capture antibody was coated onto a microtiter plate at a concentration of 4 ⁇ g/ml overnight at 4°C. The plate was washed three times with PBST and blocked with 5% BSA for 1 h. A 100 ⁇ l of the test sample (which can be diluted 2-3 fold) or the control pTie2 (used to prepare the standard curve) was added and incubated at room temperature for 2 h. After three washes with PBST, 100 ⁇ l of HRP-conjugated anti-pTyr antibody was added and incubated at room temperature for 2 h.
  • the U87 cells (human glioma cells), which express multiple RGD integrins, were used to investigate the inhibitory effect of an antibody on hAng2/ integrin-mediated cell adhesion by measuring the number of U87 cells adhering to a microplate in the presence of the antibody by Celltiter (Promega). The results were then compared with that of the positive control antibody Nesvacumab.
  • a microtiter plate was disinfected with UV irradiation, and recombinant hAng2 (Sino Biological, Cat No. 10691-H08H) was diluted to 2.5 ⁇ g/ml, and coated onto the microtiter plate at 4°C overnight. After three washes with PBS, the plate was blocked with 10% FBS in DMEM for 1 h. After discarding the blocking solution, the antibody of the present invention was added. An IgG isotype control antibody was used as a negative control, and Nesvacumab as a positive control. The highest concentration of the antibody was 100 ⁇ g/ml, and 1: 4 serial dilution was performed with a total of six concentration points.
  • the antibody was added at 100 ⁇ l/well and incubated for 90 min; then the plate was washed three times with PBS.
  • U87 MG cells were diluted to a concentration of 1 ⁇ 10 5 cells/ml in serum-free DMEM and 100 ⁇ l was added to each well. Then the cells were incubated in a CO 2 incubator at 37°C for 30 min. The cells were washed three times with 100 ⁇ l/well of PBS and the supernatant was discarded carefully. Then CellTiter-Glo (promega, Cat. No. # G7572) was added at 100 ⁇ l/well and shaken for 2 min. The absorbance was determined on a microplate reader according to the manufacturer's instructions for uses.
  • RNAs were extracted from 1 ⁇ 10 6 /ml ARPE-19, Müller, astrocyte, HUVEC and SP2-0 cells (all available from ATCC), and the absence of Ang2 transcription in ARPE-19, Müller and astrocyte cells was verified by RT-PCR. The results are shown in the electropherogram of Figure 6 .
  • the upper electropherogram shows the Ang2 PCR band generated from amplification, and there was no transcription of Ang2 gene in ARPE-19, Müller and astrocytes except for the positive control cell HUVEC.
  • the lower electropherogram shows the amplified bands of the control gene ⁇ -actin.
  • PBS was added(200 ⁇ l/well), centrifugation was performed at 400 g for 5 min, and the supernatant was discarded; fluorescent secondary antibody PE-anti human IgG Fc (Southern Biotech) diluted at 1: 200 was added into each well and incubated at 4°C for 30 min; then PBS was added at 200 ⁇ l/well, the cells were centrifuged at 400 g for 5 min, and after the supernatant was discarded, PBS was added at 100 ⁇ l/well to resuspend the cells, and the MFI value was measured by flow cytometry.
  • PE-anti human IgG Fc Pacificn Biotech
  • Ang1 is produced by vascular pericytes and Ang2 is produced by vascular endothelial cells.
  • Ang1 and Ang2 competitively bind to TIE2 receptor expressed on endothelial cells, which triggers downstream events.
  • anti-ANG2 antibody the effect of anti-ANG2 antibody on the expression of inflammatory markers in endothelial cells in the presence of both ANG1 and ANG2 was investigated.
  • HUVEC cells were transfected by lentivirus to obtain the Tie2 overexpressing HUVEC-Tie2 cells.
  • Huvec-Tie2 cells were seeded in a 24-well or 96-well plate at a concentration of 2 ⁇ 10 5 cells/ml.
  • the medium was replaced with the following conditioned media after the Huvec-Tie2 cells were incubated for 24 h:
  • the cells were washed three times with PBS, digested with 100 ⁇ l of trypsin, and stained with the anti-CD54 antibody (Biolegend Cat No.: 353108) and anti-CD106 antibody (Biolegend Cat. No.: 305810) (diluted at 1:500, at 500 ⁇ l/well). After staining and washing, the expression of CD54 and CD106 on the cells was determined by flow cytometry.
  • Ang2 can sensitize TNF- ⁇ induced expression of CD54 and CD106 on HUVEC cells in the presence of ANG1.
  • HZ81H10.4 significantly inhibites this function of Ang2, with a potency comparable to or better than the positive control antibody Nesvacumab ( Figure 7B ).
  • ICAM-1 and VCAM-1 are annotated on uniport under GO Biological Process GO: 0002438 (Acute Inflammatory Response to Antigenic Stimuli) and are involved in the migration of leukocytes across vascular endothelium.
  • the inhibitory effect of the antibody on ICAM-1/VCAM-1 expression suggests that the antibody may be beneficial for preventing leukocyte migration across vascular endothelium and leukocyte infiltration in inflammatory tissues during acute inflammatory response.
  • Ang2/Tie2 and Ang2/integrin signaling pathways are involved in the increased vascular endothelial permeability and vascular leakage under pathological conditions.
  • hz81H10.4 molecule blocks the functions of Ang2/Tie2 and Ang2/integrin. Therefore, the permeability assay in HUVEC-Tie2 cells was used to further identify the effect of the antibody on vascular leakage.
  • HUVEC cells were transfected by lentivirus to obtain the Tie2 overexpressing HUVEC-Tie2 cells.
  • a mini-well 96-well dish with insert was used, with 300 ⁇ l of culture medium in the lower chamber.
  • Adherent HUVEC-Tie2 cells (which secretes their own Ang2) in culture were digested and resuspended to 1 ⁇ 10 7 cell/ml in culture medium, and 100 ⁇ l/well was plated in the upper chamber of the dish.
  • the medium in the lower chamber was replaced every 24 h, and the medium in the upper chamber was replaced with the following experimental medium 48 hours later:
  • Acute respiratory distress syndrome is an acute and critical disease in clinical practice characterized by severe respiratory distress and progressive respiratory failure. Its pathological features include: diffuse damage to alveolar epithelial barrier and pulmonary capillary endothelial barrier, leading to inflammatory cytokine infiltration and increased lung weight.
  • a LPS-induced acute respiratory distress syndrome model in New Zealand white rabbits was used to mimic the abnormal lung function caused by acute lung inflammatory injury in human patients; and the function of anti-ANG2 antibody in helping the recovery of alveolar-capillary barrier in individual animals with pneumonia was investigated.
  • Animal species New Zealand rabbits (2-5 months old) were purchased from Suzhou Genescience Biotechnology Co., Ltd. (laboratory animal production license No.: SCXK (Su) 2020-0002 ; laboratory animal quality certificate No.: 20200907214 ). The animals were subjected to one week of acclimation prior to the initiation of the study.
  • LPS Lipopolysaccharide
  • LPS lipopolysaccharide
  • LPS lipopolysaccharide
  • LPS lipopolysaccharide
  • LPS lipopolysaccharide
  • Intratracheal nebulization of LPS was performed as follows: animals were anesthetized by intramuscular injection of Zoletil 50 (8 mg/kg, 50 mg/mL) combined with xylazine hydrochloride (0.3 mg/kg, 20 mg/mL) and then the animals were fixed to a rabbit fixator placed at 30°. An animal anesthesia laryngoscope was used to press the animal's tongue base to expose the glottis.
  • the needle (with a blunt tip) of a liquid micronebulizer that drew a quantitative amount of LPS solution was gently inserted into the trachea, then the piston was rapidly pushed to nebulize the LPS solution into the lungs, then the needle was quickly withdrawn, and the animal was released from the fixator with the head facing upward and rotated left and right to distribute the LPS as evenly as possible throughout the lung lobes.
  • the volume of LPS to be administered was round to the nearest tenth. If the LPS volume was between two volume graduation lines, LPS was drawn according to the volume value of the upper graduation line.
  • test antibody was administered according to Table 3 below.
  • Table 3 Antibody Dosing Regimen for the Groups of Acute Respiratory Distress Syndrome Animal Model Group Dose (mg/kg) Concentration (mg/ml) Volume (mL/kg) Animal No. 1 LPS low-dose group 15 7.5 2 1, 2 0 0 2 3, 4 2 LPS medium-dose group 15 7.5 2 5, 6 0 0 2 7, 8 3 LPS high-dose group 15 7.5 2 9, 10 0 0 2 11,12 Note: The first two animals in each group were injected with the test antibody via marginal ear vein, and the last two animals were injected with sodium chloride injection via marginal ear vein.
  • Anti-hAng2 antibody hz81H10.4 Inhibites leukocyte infiltration in the alveoli
  • a tracheal cannula was inserted into the left bronchus and the cannula was fixed to the bronchus with sutures.
  • a total volume of 10 mL/kg sodium chloride injection solution was drawn using a syringe and slowly injected to fill the lungs.
  • the pulmonary alveoli were lavaged three times, with each lavage lasting about 10s, and the lavage fluids were collected in a centrifuge tube with an appropriate volume.
  • the collected lavage fluid, 2 mL per animal, was retained and centrifuged at 4 °C at about 3000 rpm for 10 min. The supernatant was discarded, and the precipitate was resuspended in 1 mL of PBS buffer for total and differential leukocyte count.
  • Anti-hAng2 antibody hz81H10.4 effectively inhibited the infiltration of leukocytes and various types of inflammatory cells in the alveoli of ARDS animals.
  • the results of the LPS low-dose group are presented in Figure 9 .
  • the total white blood cell count, as well as the differential count (counts of inflammatory cells, neutrophils, lymphocytes and monocytes) in the pulmonary alveolar lavage fluid were significantly decreased compared to the control group to which the sodium chloride was injected.
  • Anti-hAng2 Antibody hz81H10.4 Improves Lung Weight Index
  • All animals in each group were dissected and the tissues were preserved.
  • the animals were placed in a refrigerator at 2-8°C after being found dead during non-working hours and dissected as soon as possible within 24 hours.
  • Figure 10 shows the calculated results of the lung weight index.
  • the normal lung weight index of male New Zealand rabbits is about 0.3 (" Study on the Chief Organ Weight and Coefficients of Different Species of Experimental Rabbits", Chinese Journal of Comparative Medicine, December 2004, Volume 14, No. 6 ).
  • Each of the three doses of LPS induced significant increases in lung weight index in individuals of the control group.
  • Administration of the antibody showed a general tendency to reduce the LPS-induced increase in lung weight index.
  • Anti-hAng2 Antibody hz81H10.4 Improves Animal Survival Rate
  • Anti-hAng2 Antibody hz81H10.4 Improves Lung Pathology in Animals
  • the left lung was excised using scissors to collect bronchoalveolar lavage fluid.
  • the remaining right lung tissues and bronchi were fixed in 10% neutral buffered formalin solution, embedded in paraffin, sliced, prepared into sections and stained with HE to observe the pathological morphology of the lung tissues.
  • Pathological examination of lung tissues was performed using a 4-point scale (minimal, mild, moderate, severe) according to the Standard Terminology for Diagnosis and Classification (DOI: 10.1177/0192623318761348 ).
  • Figure 12 shows the staining results of pathological lung sections of the ARDS model.
  • hz81H10.4 treatment can ameliorate LPS-induced alveolar/interstitial/bronchial/pleural inflammatory cell infiltration, edema/fibrin-like material exudation, lung abscess, vascular/bronchial/alveolar/interstitial degeneration/necrosis, alveolar wall thickening, thrombus and pleural hemorrhage.
  • the LPS-induced ARDS animals were treated with low dose (5 mg/kg), medium dose (15 mg/kg) and high dose (25 mg/kg) of HZ81H10.4 antibody in this Example.
  • model control group Ninety-six male animals that passed quarantine were weighed and randomly divided into model control group (9 animals, injected with sodium chloride solution), low-dose antibody test product group (7 animals), medium-dose antibody test product group (7 animals), high-dose antibody test product group (7 animals) and positive control group (6 animals, injected with dexamethasone at 2 mg/kg). The average body weights were not different across the groups.
  • the modeling agent LPS was drawn at an amount according to the body weight of the animals, and was delivered to the animals via intraperitoneal injection at 0.2 mg/kg (0.2 mL/kg), and 16 hours ( ⁇ 30 min) after the intraperitoneal injection, delivered to the animals via intratracheal nebulization at 0.5 mg/kg (0.5 mL/kg) .
  • mice were treated with drugs (one dose), intravenously in Groups 1-4, and intraperitoneallyin Gourp 5.
  • drugs one dose
  • intraperitoneal injection the model animals were subjected to CT examination and blood gas analysis and then euthanized; the bronchoalveolar lavage fluid was collected and analyzed, and the lung tissues were embedded for preparation of pathological sections.
  • Table 4 Antibody Dosing with Gradient Concentrations for the Groups of Acute Respiratory Distress Syndrome Animal Model Group Dose (mg/kg) Concentration (mg/ml) Volume (mL/kg) Animal No.
  • Anti-hAng2 Antibody Inhibits Leukocyte Infiltration in the alveoli
  • the bronchoalveolar lavage fluid was detected as in Example 12.1 to reveal leukocyte infiltration in the alveoli of ARDS animals.
  • the results are shown in Figure 13 .
  • administration of antibody hz81H10.4 significantly inhibited leukocyte infiltration in the alveoli of ARDS animals ( ⁇ : p ⁇ 0.05; ⁇ : p ⁇ 0.01; ⁇ : p ⁇ 0.001) ; and administration of antibody hz81H10.4 exhibited dose-dependent effects, with the greatest therapeutic efficacy at 25 mg/ml in improving the infiltration of leukocytes and various types of inflammatory cells (neutrophils, lymphocytes and monocytes) in the alveoli, and demonstrated a therapeutic efficacy that was comparable to that of dexamethasone in reducing the total leukocyte count and the neutrophil count, and superior to that of dexamethasone in reducing the infiltration of lymphocytes and monocytes.
  • Anti-hAng2 Antibody Enhances Oxygen Saturation in the model Animals
  • the effect of antibody on oxygen saturation of the model animals was investigated by blood gas detection.
  • the animals were anesthetized, and a midline ventral incision was performed to isolate the abdominal aorta, and about 0.5 mL of arterial blood was collected using an arterial blood collection syringe.
  • the needle was gently inserted into the blood entry port on the test card and the blood was slowly injected to fill the sample collection tube.
  • sample collection was stopped, the cap was closed, and the blood automatically entered the test tube.
  • the test card was inserted into the i-STAT ® 1 Handheld Blood Gas Analyzer (300-G. Abott Point of Care Inc. USA), and the dection indexes included PO 2 (mmHg), PCO 2 (mmHg), pH and sO 2 %.
  • the detection results of PO 2 are shown in Figure 14 .
  • ARDS model animals showed hypoxemia, and the partial pressure of oxygen was lower than 80 mmHg.
  • the antibody treated group the antibody induced improvement in oxygen saturation in ARDS model animals in a dose-dependent manner, and the improvement was statistically significant at the doses of 15 mg/ml and 25 mg/ml ( ⁇ : p ⁇ 0.05; ⁇ : p ⁇ 0.01; ⁇ : p ⁇ 0.001); moreover, the effect of the antibody dosed at 25 mg/ml was comparable to that of dexamethasone in terms of improving oxygen saturation in the model animals.
  • Anti-hAng2 Antibody Improves lung Pathology in Animals
  • the left lung was excised using scissors to collect bronchoalveolar lavage fluid.
  • the remaining right lung tissues and bronchi were fixed in 10% neutral buffered formalin solution, embedded in paraffin, sliced, prepared into sections, and stained with HE to observe the pathological morphology of the lung tissues.
  • Pathological examination of lung tissues was performed using a 4-point scale (minimal, mild, moderate, severe) according to the Standard Terminology for Diagnosis and Classification.
  • Figure 15 shows the staining results of lung pathological sections of the ARDS animals.
  • Figure 16 shows the proportion of individual animals with different grades of lung tissue inflammatory lesions in different treatment groups.
  • lung tissues of animals in the control group were observed to have mild inflammatory cell infiltration in blood vessels/alveoli/peribronchus/bronchial epithelium/interstitium/pleura, alveolar wall thickening, mild alveolar/bronchial/interstitial degeneration/necrosis, slight pleural fibrosis, and mild exudation of fibrin like material in the alveoli.
  • the administration of low-, medium- and high-doses of hz81H10.4 and positive control dexamethasone exhibited alleviative effect on LPS-induced alveolar/peribronchial/interstitial inflammatory cell infiltration, alveolar wall thickening, exudation of fibrin like material in the alveoli, alveolar wall/bronchial/interstitial degeneration/necrosis, pleural fibrosis, arterial thrombotic vasculitis and pleural inflammation.
  • Anti-hAng2 Antibody Improves CT images in Animals
  • CT images showed that the CT symptoms of local pulmonary inflammation, increased markings in both lungs, and low transmittance in both lungs wereimproved in the hz81H10.4 group.
  • LPS was administered (0.2 mg/kg intraperitoneally + 0.5 mg/kg intratracheally) to the rabbit model animals, followed by the administration of hz81H10.4 antibody (5 mg/kg intravenously).
  • the method of animal modeling and subsequent drug administration was the same as that in Example 12.2.
  • sodium chloride injection as the negative control in place of the antibody was administered intravenously, and dexamethasone (2mg/kg) as the positive control was administered intraperitoneally. There were 7 animals in each group. After antibody administration, the animals were dissected on Day 7 to calculate the lung weight index and survival rate.
  • Anti-hAng2 Antibody Improves Lung Weight Index
  • the lung weight index of the model animals was calculated to investigate the ameliorative effect of the antibody on lung weight index of the ARDS animals. Compared with the control group, a significant improvement in lung weight index was observed in the antibody group.
  • Anti-hAng2 Antibody Improves Survival Rate in Animals
  • mice of the IBI304 group and IBI304 + 81H10.4 group were intravitreously injected in both eyes, with 0.6 mg/mL IBI304 and 0.6 mg/mL IBI304 +1.8 mg/mL hz81H10.4 at 30 ⁇ g/eye and 30 +90 ⁇ g/eye, respectively.
  • the animals in the model control group were injected with an equal volume of 0.9% sodium chloride injection.
  • Color fundus photography and fundus fluorescein angiography were performed on Day 7, Day14, Day 21 and Day 28 post drug administration to observe the inhibitory effect of the test products on choroidal neovascularization.
  • the animals were euthanized 29 days after drug administration, and their two eyes were excised for histological examination with HE staining.
  • fluorescein leakage area fluorescein leakage area before drug administration ⁇ relative fluorescence intensity ⁇ fluorescein leakage area after drug administration ⁇ relative fluorescence intensity / fluorescein leakage area before drug administration ⁇ relative fluorescence intensity
  • Relative fluorescence intensity mean fluorescence intesity in fluorescence spot leakage area ⁇ mean fluorescence intensity in the background area
  • the grading scale of the light spots captured by fluorescence angiography after modeling is as follow: Grade 1: the light spot does not have high-intensity fluorescence; Grade 2: the light spot has high-intensity fluorescence but without fluorescein leakage; Grade 3: the light spot has high-intensity fluorescence with mild fluorescein leakage that does not extend beyond the edge of the light spot; Grade 4: the light spot has high-intensity fluorescence, with significant fluorescein leakage beyond the edge of the light spot. The percentage of light spots of grade 2-4 was counted.
  • rhesus monkeys were anesthetized with sodium pentobarbital according to body weight (intravenous injection of about 30 mg/kg, and the dose could be adjusted according to animal health conditions), euthanized by exsanguination via the abdominal aorta or femoral artery, macroscopic observation was performed, and bilateral eyeballs were excised.
  • the laser-injuryed sites were histologically examined by HE staining to detect the areas with tissue hyperplasia, edema and retinal detachment and the sizes of injured and swollen areas were calculated by the Image J software.
  • the results showed that in the control group, there was large areas of edema at the laser-induced modeling sites, accompanied with significantly irregular retina morphology and tissue proliferation; after treatment with IBI304, edema was significantly ameliorated; in addition, a further improvement in retinal morphology and tissue proliferation in the laser-induced modeling sites was observed in the IBI304 + hz81H10.4 treatment group over the IBI304 monotherapy.
  • the results are shown in Figure 21 .
  • the results from the pathological sections reflect that the combination of the antibody of the present invention with anti-VEGF agent exhibited a superior effect in improving retinal morphology, inhibiting retinal choroidal neovascularization, and enhancing vascular integrity compared with anti-VEGF monotherapy.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
EP21827966.9A 2020-06-22 2021-06-21 Anti-ang-2-antikörper und verwendung davon Pending EP4183802A1 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN202010573625 2020-06-22
CN202011428549 2020-12-09
PCT/CN2021/101243 WO2021259200A1 (zh) 2020-06-22 2021-06-21 抗ang-2抗体及其用途

Publications (1)

Publication Number Publication Date
EP4183802A1 true EP4183802A1 (de) 2023-05-24

Family

ID=79281955

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21827966.9A Pending EP4183802A1 (de) 2020-06-22 2021-06-21 Anti-ang-2-antikörper und verwendung davon

Country Status (6)

Country Link
EP (1) EP4183802A1 (de)
JP (1) JP2023530187A (de)
CN (1) CN115734971A (de)
AU (1) AU2021294703A1 (de)
CA (1) CA3190173A1 (de)
WO (1) WO2021259200A1 (de)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116003591B (zh) * 2022-12-12 2023-09-19 三门峡市眼科医院 Ang-2抗体及其应用

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0154316B1 (de) 1984-03-06 1989-09-13 Takeda Chemical Industries, Ltd. Chemisch modifizierte Lymphokine und Verfahren zu ihrer Herstellung
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
DE68925966T2 (de) 1988-12-22 1996-08-29 Kirin Amgen Inc Chemisch modifizierte granulocytenkolonie erregender faktor
ATE375365T1 (de) 1998-04-02 2007-10-15 Genentech Inc Antikörper varianten und fragmente davon
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US7087411B2 (en) 1999-06-08 2006-08-08 Regeneron Pharmaceuticals, Inc. Fusion protein capable of binding VEGF
AU2005319382B2 (en) * 2004-12-21 2011-04-07 Astrazeneca Ab Antibodies directed to angiopoietin-2 and uses thereof
US8133979B2 (en) * 2008-12-16 2012-03-13 Hoffmann-La Roche Inc. Antibodies against human angiopoietin 2
US9994632B2 (en) * 2014-05-26 2018-06-12 Samsung Electronics Co., Ltd. Humanized or affinity-matured anti Ang-2 antibody and uses thereof
KR20170082594A (ko) 2014-11-10 2017-07-14 에프. 호프만-라 로슈 아게 항-ang2 항체 및 사용 방법
US20160144025A1 (en) * 2014-11-25 2016-05-26 Regeneron Pharmaceuticals, Inc. Methods and formulations for treating vascular eye diseases
US10836819B2 (en) * 2016-08-23 2020-11-17 Medimmune Limited Anti-VEGF-A and anti-ANG2 antibodies and uses thereof

Also Published As

Publication number Publication date
JP2023530187A (ja) 2023-07-13
CA3190173A1 (en) 2021-12-30
AU2021294703A1 (en) 2023-02-23
CN115734971A (zh) 2023-03-03
WO2021259200A1 (zh) 2021-12-30

Similar Documents

Publication Publication Date Title
US11248052B2 (en) Antigen binding proteins that bind to a complex comprising β-Klotho and an FGF receptor
JP7065822B2 (ja) β‐クロトー、FGF受容体、およびその複合体と結合するヒト抗原結合タンパク質
US11370837B2 (en) Anti-TIGIT antibody and use thereof
RU2346701C2 (ru) ХИМЕРНЫЕ И ГУМАНИЗИРОВАННЫЕ АНТИТЕЛА ПРОТИВ ИНТЕГРИНА α5β1, КОТОРЫЕ МОДУЛИРУЮТ АНГИОГЕНЕЗ
CA2705923A1 (en) Antibodies to lrp6
US11897965B2 (en) Anti-Jagged1 antigen binding proteins
CA2904337A1 (en) Chrdl-1 antigen binding proteins and methods of treatment
EP4169947A1 (de) Anti-claudin18.2-antikörper und verwendung davon
EP4183802A1 (de) Anti-ang-2-antikörper und verwendung davon
US20230399390A1 (en) Novel anti-nogo-a antibodies
WO2020243443A1 (en) Alk7 binding proteins and uses thereof
US20220127344A1 (en) Anti-sema3a antibodies and their uses for treating a thrombotic disease of the retina
CN114591428A (zh) 抗Dsg1抗体及其应用
EP1951759B1 (de) Anti-egfr-antikörper

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20230119

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)